MedPath

NTLA-2001 Advanced Drug Monograph

Published:Oct 4, 2025

Generic Name

NTLA-2001

Drug Type

Biotech

NTLA-2001 (Nexiguran Ziclumeran): A Comprehensive Clinical and Scientific Review of a CRISPR-Based Therapy for Transthyretin Amyloidosis

I. Executive Summary

NTLA-2001, also known by the non-proprietary name nexiguran ziclumeran (nex-z), is a first-in-class, investigational gene-editing therapeutic agent designed to address the underlying cause of transthyretin amyloidosis (ATTR). Developed through a collaboration between Intellia Therapeutics and Regeneron Pharmaceuticals, NTLA-2001 leverages the Nobel Prize-winning CRISPR/Cas9 technology to achieve permanent inactivation of the transthyretin (TTR) gene within hepatocytes.[1] The therapy is administered as a single intravenous infusion, with the objective of providing a "one-and-done," potentially curative treatment for this rare, progressive, and fatal disease.[3] By permanently halting the production of the pathogenic TTR protein, NTLA-2001 offers the possibility of stopping and even reversing the debilitating complications of ATTR amyloidosis.[5]

The foundational clinical evidence for NTLA-2001 comes from a landmark Phase 1 trial (NCT04601051), which demonstrated unprecedented pharmacodynamic effects in patients with both polyneuropathy (ATTRv-PN) and cardiomyopathy (ATTR-CM) manifestations of the disease. A single administration of NTLA-2001 resulted in rapid, dose-dependent reductions in serum TTR protein concentrations. At the optimal dose levels, patients achieved mean TTR reductions exceeding 90%, an effect that has proven to be remarkably durable, with sustained suppression observed in follow-up periods extending beyond two years.[5] These profound and lasting reductions in the disease-causing protein have been accompanied by encouraging signs of clinical disease stabilization and improvement across key neurological and cardiac endpoints.[8] The therapy has been generally well tolerated, with a manageable safety profile characterized primarily by mild, transient infusion-related reactions.[7]

The success of the Phase 1 study has propelled NTLA-2001 into late-stage clinical development, making it the first systemically administered in vivo CRISPR-based candidate to advance to pivotal trials.[4] The comprehensive Phase 3 program includes the MAGNITUDE study (NCT06128629) for patients with ATTR-CM and the planned MAGNITUDE-2 study (NCT06672237) for patients with ATTRv-PN.[1] The ambitious design of these trials, particularly the use of cardiovascular mortality and events as a primary endpoint in the MAGNITUDE study, underscores the high level of confidence in the therapy's potential to deliver transformative clinical outcomes.[2]

NTLA-2001 represents a potential paradigm shift in the field of genetic medicine. Its mechanism moves beyond the chronic management model of existing TTR stabilizers and gene silencers, introducing the concept of a single-administration treatment that permanently eliminates the source of the pathology. If the robust efficacy and favorable safety profile are confirmed in pivotal trials, NTLA-2001 could fundamentally reset the standard of care for ATTR amyloidosis and validate the systemic CRISPR/Cas9-LNP platform as a powerful new modality for treating a host of other genetic diseases.

II. The Clinical Challenge of Transthyretin Amyloidosis (ATTR)

A. Pathophysiology of TTR Misfolding and Amyloid Deposition

Transthyretin (TTR), historically known as prealbumin, is a 127-amino acid protein synthesized predominantly by the liver (>95%), with minor production in the choroid plexus of the brain and the retinal pigment epithelium.[11] In its physiological state, TTR circulates in plasma and cerebrospinal fluid as a stable, soluble homotetramer, formed by the assembly of four identical monomeric subunits.[12] Its primary biological functions are to serve as a transporter for thyroxine (a thyroid hormone) and for retinol (vitamin A) via its binding to retinol-binding protein.[11]

The pathophysiology of ATTR amyloidosis is rooted in a process of protein misfolding known as the amyloidogenic cascade.[15] The critical, rate-limiting step in this cascade is the dissociation of the stable TTR tetramer into its constituent monomers.[13] Once dissociated, these monomers are conformationally unstable and prone to misfolding. The misfolded monomers then self-assemble into insoluble, highly organized aggregates with a characteristic cross-β sheet structure, known as amyloid fibrils.[11] These fibrils progressively deposit in the extracellular space of various organs and tissues, disrupting their normal architecture and function.[3] The continuous accumulation of these amyloid deposits leads to progressive organ damage, culminating in organ failure and death if left untreated.[19]

B. The Clinical Spectrum: Hereditary (ATTRv) vs. Wild-Type (ATTRwt) Disease

The instability of the TTR tetramer that initiates the amyloidogenic cascade arises from two distinct etiologies, defining the two major forms of the disease: hereditary and wild-type ATTR.

Hereditary ATTR (ATTRv) is an autosomal dominant genetic disorder caused by mutations in the TTR gene, which is located on chromosome 18.[11] An affected individual inherits one copy of the altered gene from a parent and has a 50% probability of passing it on to each of their offspring.[12] More than 120 different pathogenic point mutations have been identified.[11] These mutations result in single amino acid substitutions that structurally destabilize the TTR protein, thereby accelerating tetramer dissociation and promoting amyloid fibril formation.[11] The clinical presentation, age of onset, and pattern of organ involvement can vary significantly depending on the specific mutation and the patient's geographic origin.[21] Globally, an estimated 50,000 people are living with ATTRv amyloidosis.[1]

Wild-Type ATTR (ATTRwt), previously referred to as senile systemic amyloidosis, occurs in individuals who do not have a mutation in their TTR gene.[1] In this form of the disease, the normal, non-mutated TTR protein becomes conformationally unstable as a consequence of the aging process.[17] While the exact triggers are not fully understood, this age-related instability leads to the same pathological cascade of tetramer dissociation, misfolding, and amyloid deposition.[23] ATTRwt primarily affects the heart and is most commonly diagnosed in men over the age of 60.[23] It is now recognized as a significantly underdiagnosed cause of heart failure in the elderly and is far more prevalent than the hereditary form, with an estimated 200,000 to 500,000 people affected worldwide.[1]

The existence of these two distinct disease mechanisms—one driven by an inherited genetic flaw and the other by an age-related process—necessitates a therapeutic approach that can address the final common pathway of TTR misfolding, regardless of its origin. A strategy like gene knockout, which targets the production of the TTR protein itself, provides a universal solution applicable to all forms of ATTR. By eliminating the source protein, such a therapy is agnostic to whether the TTR instability is caused by a specific mutation or by the aging process, making it a potentially definitive treatment for the entire patient population.

C. Dominant Phenotypes: Cardiomyopathy (ATTR-CM) and Polyneuropathy (ATTRv-PN)

While ATTR is a systemic disease, its clinical manifestations typically cluster into two predominant, though often overlapping, phenotypes: cardiomyopathy and polyneuropathy.

ATTR Cardiomyopathy (ATTR-CM) is characterized by the infiltration of amyloid fibrils into the myocardium.[27] This deposition occupies the interstitial spaces, causing the ventricular walls to become progressively thickened and rigid.[3] The resulting restrictive cardiomyopathy impairs the heart's ability to relax and fill with blood, leading to diastolic dysfunction and, in advanced stages, systolic dysfunction and heart failure.[3] Patients commonly experience symptoms such as shortness of breath, fatigue, swelling in the lower extremities (edema), and cardiac arrhythmias.[19] ATTR-CM is the principal manifestation of ATTRwt and is also a feature of certain hereditary variants, such as the V122I mutation, which is more common in individuals of African descent.[12] The prognosis is poor, with a median survival of 2 to 6 years after diagnosis without effective treatment.[19]

Hereditary ATTR Polyneuropathy (ATTRv-PN) results from amyloid deposition in the peripheral and autonomic nervous systems.[12] This leads to a progressive, length-dependent sensorimotor polyneuropathy, characterized by symptoms such as numbness, tingling, burning pain, and muscle weakness, typically starting in the lower extremities.[12] Concurrently, autonomic neuropathy causes dysfunction in involuntary bodily processes, leading to orthostatic hypotension (a sharp drop in blood pressure upon standing), gastrointestinal issues (diarrhea, constipation), urinary problems, and erectile dysfunction.[12] The disease typically progresses through defined stages, from mild sensory symptoms with preserved walking ability (Stage I) to severe disability requiring a wheelchair or confinement to bed (Stage III).[11] ATTRv-PN is the classic presentation for several

TTR mutations, most notably the Val30Met (V30M) variant.[11]

The clinical presentation of ATTR is often a significant diagnostic challenge. Its multisystemic nature and the staggered onset of seemingly unrelated symptoms—such as carpal tunnel syndrome, spinal stenosis, heart failure, and peripheral neuropathy—can mimic more common conditions.[18] This frequently leads to misdiagnosis and a substantial delay, averaging four years from symptom onset to an accurate diagnosis.[18] This delay has profound clinical implications. By the time ATTR is correctly identified, patients often have advanced disease with significant, and in many cases irreversible, organ damage. This reality creates a critical unmet need for therapies that are not merely disease-modifying but are powerful enough to halt disease progression and potentially reverse damage, even when initiated at later stages.

D. Current Therapeutic Landscape and Unmet Medical Needs

The modern therapeutic landscape for ATTR amyloidosis is centered on two primary strategies: stabilizing the TTR protein to prevent its misfolding or silencing the TTR gene to reduce its production.[5]

TTR Stabilizers: These small-molecule drugs bind to the thyroxine-binding sites on the TTR tetramer, kinetically stabilizing its structure and preventing its dissociation into amyloidogenic monomers.[33]

Tafamidis (marketed as Vyndaqel and Vyndamax) and the more recently approved acoramidis (Attruby) are the leading agents in this class, approved for the treatment of ATTR-CM.[35] They have been shown to reduce cardiovascular mortality and hospitalization and slow the decline in functional capacity.[34]

Gene Silencers: This class of drugs utilizes RNA-based technologies to reduce the hepatic synthesis of TTR protein. They work by targeting the TTR messenger RNA (mRNA) for degradation, thereby lowering the levels of both mutant and wild-type TTR in circulation.[5] This class includes:

  • Small interfering RNA (siRNA) therapies: Patisiran (Onpattro), administered via intravenous infusion every three weeks, and vutrisiran (Amvuttra), a subcutaneously injected therapy given every three months.[36]
  • Antisense oligonucleotide (ASO) therapies: Inotersen (Tegsedi) and eplontersen (Wainua), both administered via subcutaneous injection.[36]

These agents are approved for treating the polyneuropathy of hATTR, with vutrisiran also recently gaining approval for ATTR-CM.36 They have demonstrated the ability to halt or improve neurological impairment.42

Despite these significant therapeutic advances, critical unmet needs persist. All current treatments require chronic, lifelong administration—whether daily oral pills, frequent subcutaneous injections, or regular intravenous infusions.[5] This imposes a substantial treatment burden on patients and healthcare systems. Furthermore, while these therapies can slow disease progression, they do not offer a cure, and patients may continue to experience disease advancement over time.[5] This context highlights the need for a novel therapeutic approach that can provide a more durable, and ideally permanent, effect from a more convenient administration regimen, thereby offering the potential to definitively halt the disease.[3]

III. NTLA-2001: A Novel In Vivo Gene-Editing Therapeutic

NTLA-2001 represents a fundamentally new approach to treating ATTR amyloidosis, moving beyond disease management to direct intervention at the genomic level. It is the first therapy based on the Nobel Prize-winning CRISPR/Cas9 technology to be administered systemically for the purpose of editing genes in vivo, or directly inside the human body.[1]

A. Core Technology: CRISPR/Cas9-Mediated Gene Knockout

The therapeutic engine of NTLA-2001 is a two-part genome editing system designed not to correct the TTR gene, but to permanently inactivate, or "knock out," its function.[3] The two active components are:

  1. A human-optimized messenger RNA (mRNA) molecule, approximately 4,400 nucleotides in length, that encodes the Streptococcus pyogenes Cas9 (SpyCas9) protein. The Cas9 protein is an endonuclease, an enzyme that functions as a pair of "molecular scissors" capable of cutting DNA.[3]
  2. A single guide RNA (sgRNA), a molecule of approximately 35 kDa, which is engineered to be complementary to a specific target sequence within the human TTR gene. The sgRNA acts as a molecular guide, directing the Cas9 enzyme to its precise location on the genome.[3]

The therapeutic goal is to induce a permanent loss of function in the TTR gene. After the Cas9 endonuclease is guided to the target site by the sgRNA, it creates a double-strand break in the DNA. The cell's natural DNA repair machinery, primarily through a process called non-homologous end joining (NHEJ), attempts to repair this break. NHEJ is an error-prone process that often results in the insertion or deletion of a small number of nucleotides, known as "indels," at the site of the break. These indels disrupt the gene's coding sequence and reading frame, permanently preventing the cell from transcribing and translating the gene into a functional TTR protein.[3] This mechanism effectively and irreversibly shuts down the production of the disease-causing protein at its source.

This approach represents a fundamental shift from "disease modification" to "source elimination." Whereas TTR stabilizers manage the protein after it has been produced and RNA silencers require continuous intervention to suppress mRNA translation, NTLA-2001 operates at the most fundamental level of biology by altering the genomic DNA itself. By permanently removing the genetic blueprint for the pathogenic protein, this single intervention offers the potential for a definitive, lifelong therapeutic effect, embodying the concept of a "one-and-done" cure.[1]

B. Detailed Mechanism of Action: From Intravenous Infusion to Permanent TTR Gene Inactivation

The clinical application of NTLA-2001 involves a multi-step biological process that begins with administration and culminates in precise gene editing within the nucleus of liver cells.

  1. Administration and Systemic Circulation: NTLA-2001 is administered as a single dose via intravenous (IV) infusion.[3] Upon entering the bloodstream, the lipid nanoparticle (LNP) delivery vehicle is rapidly coated with endogenous proteins, most importantly Apolipoprotein E (ApoE).[3]
  2. Targeted Delivery to the Liver: This ApoE coating serves as a natural targeting ligand. Hepatocytes (liver cells) are rich in low-density lipoprotein (LDL) receptors, which recognize and bind to ApoE. This interaction facilitates the selective and preferential uptake of the LNPs by the liver, the primary site of TTR synthesis, while minimizing exposure to other tissues.[3]
  3. Cellular Uptake and Endosomal Release: Following binding to the LDL receptor, the LNP is internalized into the hepatocyte through endocytosis, becoming enclosed within a vesicle called an endosome.[3] The proprietary ionizable lipids within the LNP are designed to interact with the acidic environment of the endosome, leading to the disruption of the endosomal membrane and the release of the LNP's cargo—the Cas9 mRNA and the TTR-targeting sgRNA—into the cell's cytoplasm.[3]
  4. Execution of Gene Editing: Once in the cytoplasm, the cell's natural translational machinery reads the Cas9 mRNA and synthesizes the Cas9 protein. This newly formed Cas9 protein then binds with the co-delivered sgRNA to form a ribonucleoprotein (RNP) complex. This complex is the active gene-editing machinery. It is then transported into the cell's nucleus, where the sgRNA guides the Cas9 enzyme to the specific, complementary DNA sequence within the TTR gene. Upon successful binding, the Cas9 endonuclease cleaves both strands of the DNA, initiating the permanent gene knockout process described previously.[3]

C. The Role of Lipid Nanoparticle (LNP) Delivery in Hepatocyte Targeting

The delivery vehicle is as critical to the success of NTLA-2001 as the CRISPR/Cas9 payload itself. Intellia's proprietary non-viral platform utilizes a carefully formulated LNP to encapsulate, protect, and deliver the RNA components to the target cells.[4]

Composition: The LNP is a multi-component system composed of a proprietary ionizable lipid (which facilitates endosomal escape), a phospholipid (providing structural integrity), cholesterol (stabilizing the particle), and a PEGylated lipid (a lipid attached to polyethylene glycol, which helps to shield the particle in circulation and control its size).[3] This formulation is designed to safely carry the large Cas9 mRNA and the sgRNA through the bloodstream to the liver.

Advantages of LNP Delivery: The selection of a non-viral LNP system over viral vectors, such as adeno-associated viruses (AAVs), is a critical strategic decision that confers significant advantages in safety and platform versatility.

  • Reduced Immunogenicity: Unlike viral vectors, which can trigger strong and persistent immune responses, LNPs are generally associated with a more limited and transient immune reaction. This is crucial for patient safety and reduces the risk of the body developing immunity that would preclude future treatments.[5]
  • Transient Nature and Non-Integration: The LNP and its RNA cargo are transient. After delivering the editing machinery, the components are naturally metabolized and cleared from the body. The CRISPR/Cas9 system itself does not integrate into the host genome, mitigating the theoretical risk of insertional mutagenesis (disruption of other essential genes) that can be associated with some viral vectors.[5]
  • Redosing Capability: The transient nature and lower immunogenicity of the LNP platform enable the theoretical possibility of redosing. This was clinically validated in a small cohort of patients in the Phase 1 trial. Three patients who received an initial low dose were successfully re-dosed two years later with a higher dose, achieving the desired level of TTR reduction.[1] While Intellia has stated that redosing is not planned for the ATTR program, as a single high dose appears profoundly effective, this demonstration is a landmark achievement. It de-risks the entire therapeutic platform by proving that an initial suboptimal response does not foreclose the possibility of future therapeutic success. This capability is a vital asset for the development of future therapies for other genetic diseases where an additive effect or dose titration may be desirable.[46]

IV. Clinical Development Program

The clinical development of NTLA-2001 is a joint effort, guided by a methodical progression from early-phase dose-finding to a comprehensive, late-stage program designed to establish its efficacy and safety across the full spectrum of ATTR amyloidosis.

A. Overview of the Intellia Therapeutics and Regeneron Collaboration

NTLA-2001 is being co-developed by Intellia Therapeutics and Regeneron Pharmaceuticals under a broad strategic collaboration agreement established in 2016.[1] Within this partnership, Intellia serves as the lead party responsible for the global development and subsequent commercialization of the NTLA-2001 program.[1] This collaboration combines Intellia's leadership in CRISPR-based gene editing with Regeneron's extensive experience in drug development and genetics, creating a powerful synergy that has accelerated the program's progress.[7]

B. Phase 1 Study (NCT04601051) Design and Population

The first-in-human study of NTLA-2001 was a foundational trial designed to rigorously assess the therapy's safety and establish its pharmacodynamic effect.

Study ParameterDescription
Official TitleStudy to Evaluate Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of NTLA-2001 in Patients With Hereditary Transthyretin Amyloidosis With Polyneuropathy (ATTRv-PN) and Patients With Transthyretin Amyloidosis-Related Cardiomyopathy (ATTR-CM) 49
Phase1 49
DesignOpen-label, multi-center, two-part study. Part 1 was a single-ascending dose escalation phase, and Part 2 was a single-dose expansion cohort at the selected dose.49
Patient PopulationAdults aged 18 to 80 years with a confirmed diagnosis of either ATTRv-PN or ATTR-CM (both hereditary and wild-type forms were included).49
InterventionA single intravenous (IV) infusion of NTLA-2001.49
Dose Cohorts (Part 1)Four dose levels were evaluated in the polyneuropathy arm: 0.1 mg/kg, 0.3 mg/kg, 0.7 mg/kg, and 1.0 mg/kg.10 Two dose levels were evaluated in the cardiomyopathy arm: 0.7 mg/kg and 1.0 mg/kg.50
Primary ObjectivesTo assess the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of NTLA-2001. The primary PD endpoint was the change from baseline in serum TTR protein concentration.49

The study enrolled patients across sites in the U.K. and New Zealand, and later expanded globally.[48] Key inclusion criteria for the cardiomyopathy arm included patients with New York Heart Association (NYHA) Class I-III heart failure and a minimum functional capacity demonstrated by the ability to walk at least 150 meters on the 6-minute walk test (6-MWT).[49] The trial is now closed to new enrollment, having successfully identified an optimal dose and provided the foundational data for the pivotal Phase 3 program.[4]

C. Pivotal Phase 3 Program: The MAGNITUDE and MAGNITUDE-2 Trials

Based on the compelling results from the Phase 1 study, Intellia has launched a comprehensive Phase 3 program to definitively evaluate NTLA-2001 (nex-z) in both major phenotypes of ATTR amyloidosis. This dual-pronged strategy is designed to maximize the therapy's potential market and address the needs of the entire patient population.

MAGNITUDE (NCT06128629) for ATTR-CM:

This is the pivotal trial for the larger cardiomyopathy patient population.

  • Design: A global, multinational, randomized, double-blind, placebo-controlled study.[2]
  • Population and Dosing: The study aims to enroll approximately 765 adult patients with either hereditary or wild-type ATTR-CM. Participants are randomized in a 2:1 ratio to receive either a single 55 mg fixed-dose infusion of NTLA-2001 or a placebo.[2] The 55 mg dose was selected based on Phase 1 data and pharmacokinetic modeling, which indicated it provides a similar drug exposure to the highly effective 1.0 mg/kg weight-based dose.[1]
  • Primary Endpoint: The trial is designed to assess clinical outcomes, with a primary endpoint defined as a composite of cardiovascular-related mortality and cardiovascular-related events.[2] This choice of a "hard" clinical endpoint, rather than a surrogate biomarker, reflects a high degree of confidence in the therapy's ability to translate profound TTR reduction into meaningful improvements in patient survival and hospitalization rates. Successfully meeting this endpoint would provide unequivocal evidence of clinical benefit and would likely establish NTLA-2001 as the definitive standard of care.
  • Status: The U.S. FDA cleared the Investigational New Drug (IND) application for this trial in October 2023.[4] The first patient was dosed in March 2024, and the trial is actively enrolling patients at over 35 sites across more than 12 countries.[2]

MAGNITUDE-2 (NCT06672237) for ATTRv-PN:

This trial is tailored to the specific needs of the hereditary polyneuropathy patient population.

  • Design: A randomized, double-blind, placebo-controlled study.[8]
  • Population and Dosing: The study is planned as a smaller, more focused trial involving approximately 50 adult patients with ATTRv-PN, conducted primarily at sites outside the U.S. Participants will be randomized 1:1 to receive a single 55 mg infusion of nex-z or placebo.[8]
  • Primary Endpoints: The co-primary endpoints are the change from baseline in the modified Neuropathy Impairment Score +7 (mNIS+7), a validated measure of neurological function, at 18 months, and the change in serum TTR concentration at day 29.[8]
  • Status: Intellia plans to dose the first patient in the MAGNITUDE-2 trial in the first quarter of 2025 and has stated an intention to submit a Biologics Licensing Application (BLA) for this indication by 2028.[56]

This parallel development strategy is both methodical and ambitious. It leverages the robust Phase 1 data to launch two distinct pivotal trials, each tailored to the specific patient population and regulatory endpoints relevant to the two major disease phenotypes. This comprehensive approach positions NTLA-2001 to potentially secure a broad label covering the full spectrum of ATTR amyloidosis.

V. Analysis of Clinical Efficacy

The clinical efficacy of NTLA-2001 has been primarily evaluated through its profound pharmacodynamic effect on serum TTR protein levels, with emerging data now demonstrating a corresponding positive impact on clinical manifestations of the disease.

A. Pharmacodynamic Response: Dose-Dependent Reduction of Serum TTR Protein

The Phase 1 study provided the first clinical evidence of the potent activity of NTLA-2001. Treatment resulted in rapid and dose-dependent reductions in serum TTR concentrations, with the maximal effect observed by day 28 post-infusion.[5] The dose-response relationship was clearly established in the polyneuropathy arm of the study.

Dose Level (mg/kg)Number of Patients (n)Mean % TTR Reduction at Day 28Maximum % TTR Reduction Observed
0.1352% 10-
0.3387% 1096% 48
0.7386% 10-
1.0693% 1098% 7

These results demonstrate a clear dose-response relationship, with the highest doses (0.7 mg/kg and 1.0 mg/kg) achieving mean TTR reductions of approximately 90% or greater. This level of TTR knockdown is not just statistically significant but biologically profound. It is hypothesized that reducing circulating TTR protein to such a low level may fall below a critical threshold required to sustain the formation of new amyloid fibrils, thereby halting disease progression and potentially allowing the body's natural clearance mechanisms to begin resolving existing deposits.[6] This level of reduction is greater than that typically reported for existing gene-silencing therapies, which achieve mean reductions in the range of 70-85% [38], suggesting a potential for superior clinical outcomes. In the cardiomyopathy arm, the effect was similarly potent and consistent, with mean TTR reductions of 93% and 92% observed at the 0.7 mg/kg and 1.0 mg/kg doses, respectively.[3]

B. Durability of Effect: Sustained TTR Suppression in Long-Term Follow-Up

A critical question for a "one-and-done" gene-editing therapy is the durability of its effect. The long-term follow-up data from the Phase 1 trial have been exceptionally strong, confirming that the TTR reduction achieved after a single dose is sustained over time. Initial follow-up showed that the deep reductions observed at day 28 remained stable through 6, 9, and 12 months, with no evidence of TTR levels rebounding.[5]

More recent data, presented with a cut-off of August 2024, have extended this observation period significantly. With over 25 patients having been followed for two years or more, the serum TTR reductions have remained "virtually unchanged" from the levels achieved in the first month.[8]

  • In the ATTR-CM arm, the mean serum TTR reduction was 90% at 12 months. In the 11 patients who had reached the 24-month timepoint, this profound reduction was fully sustained.[8]
  • In the ATTRv-PN arm, the mean TTR reduction was 91% at 12 months. This effect was also maintained in the 16 patients who had reached 24 months of follow-up.[8]

This remarkable durability provides strong evidence that the in vivo gene editing is permanent and leads to a lifelong suppression of TTR protein production from the liver, fulfilling the central premise of the therapy.

C. Impact on Clinical Manifestations: Neurological and Cardiac Endpoint Analysis

The ultimate measure of success is whether the profound reduction in TTR protein translates into meaningful clinical benefit for patients. Emerging data from the Phase 1 study suggest that it does, with favorable trends observed in both the cardiomyopathy and polyneuropathy cohorts.

Cardiomyopathy (ATTR-CM) Arm (n=36):

Despite enrolling a patient population with a high burden of disease (50% were NYHA Class III at baseline), treatment with NTLA-2001 led to evidence of disease stabilization or improvement at the 12-month follow-up.8

  • Cardiac Biomarkers: Key markers of cardiac stress and damage showed stability. 81% of patients had stable or improved levels of N-terminal pro-B-type natriuretic peptide (NT-proBNP), and 94% had stable or improved levels of high-sensitivity Troponin T (hs-Troponin T).[8]
  • Functional Capacity and Quality of Life: Functional status was preserved or enhanced. 77% of patients showed stability or improvement on the 6-minute walk test (6MWT), and 92% had a stable or improved NYHA functional classification.[8] Quality of life, measured by the Kansas City Cardiomyopathy Questionnaire (KCCQ), also showed a clinically meaningful median improvement.[8]
  • Composite Assessment: Strikingly, 66% of patients demonstrated stability or improvement across all three key cardiac markers (NT-proBNP, hs-Troponin T, and 6MWT), providing a strong, integrated signal of a disease-modifying effect.[8]

Polyneuropathy (ATTRv-PN) Arm:

Patients in the polyneuropathy arm also showed encouraging signs of neurological improvement and preserved quality of life over long-term follow-up.

  • Neurological Function: At the 24-month timepoint, patients demonstrated a mean improvement (a negative change indicates improvement) in the Neuropathy Impairment Score (NIS) of -4.5 to -5.2 points from baseline. The more comprehensive modified NIS+7 (mNIS+7) score showed a mean improvement of -8.5 points.[9] These results stand in contrast to the expected progressive worsening of neuropathy in untreated patients.
  • Quality of Life and Nutritional Status: The Norfolk Quality of Life–Diabetic Neuropathy (QoL-DN) score, a patient-reported outcome, improved by a mean of -8.5 points at 24 months, indicating a meaningful improvement in quality of life. Nutritional status, assessed by the modified body mass index (mBMI), also showed improvement.[9]

D. Special Considerations: Clinical Data on Redosing Capability

A unique and important aspect of the NTLA-2001 clinical program was the demonstration of its redosing capability. The first three patients enrolled in the study received the lowest dose of 0.1 mg/kg, which resulted in a suboptimal median TTR reduction of 52%.[1] After a two-year observation period, these patients were offered the opportunity to receive a second, follow-on dose of 55 mg.[1]

All three patients opted for the second dose, and the results were highly successful. After the 55 mg redosing, the patients achieved a median TTR reduction of 90% at day 28. This corresponded to a 95% median reduction from their original pre-treatment baseline levels, bringing them to the target therapeutic range seen in patients who received the optimal dose initially.[1] The redosing was generally well tolerated.[1] This experiment provided the first-ever clinical proof-of-concept for redosing an

in vivo CRISPR-based therapy. It serves as a powerful de-risking event for Intellia's entire LNP platform, demonstrating that an initial suboptimal response does not preclude achieving a full therapeutic effect with a subsequent dose, a critical advantage for the development of future genetic medicines.[46]

VI. Safety and Tolerability Profile

For a first-in-class therapeutic modality involving permanent alteration of the human genome, the safety and tolerability profile is of paramount importance. The clinical data gathered to date from the Phase 1 study of NTLA-2001 have been highly encouraging, indicating a favorable and manageable safety profile.

A. Comprehensive Review of Adverse Events from Phase 1 Studies

Across all dose levels tested and in both the polyneuropathy and cardiomyopathy patient populations, NTLA-2001 has been consistently reported as being generally well tolerated.[3] The vast majority of adverse events (AEs) reported have been mild (Grade 1) in severity. In an early analysis of the first 15 patients in the polyneuropathy arm, 73% (11 patients) reported a maximal AE severity of Grade 1.[7] The most frequently observed AEs across the study included headache, infusion-related reactions, back pain, rash, and nausea.[7]

Adverse Event ProfileSummary of Findings (Polyneuropathy Arm, N=15, as of Feb 2022)
Overall TolerabilityGenerally well tolerated at all four dose levels (0.1, 0.3, 0.7, 1.0 mg/kg).10
Maximal AE SeverityThe majority of adverse events were mild (Grade 1); reported by 73% (n=11) of patients.10
Most Frequent AEsHeadache, infusion-related reactions, back pain, rash, and nausea.10
Infusion-Related ReactionsAll were considered mild and resolved without clinical sequelae.7
Serious Adverse Events (SAEs)One possibly related Grade 3 event (vomiting) in a patient with a history of gastroparesis. One unrelated SAE of COVID-19 pneumonia.10
Liver FindingsNo clinically significant liver findings were observed.10

This safety profile is highly favorable, particularly given the novelty and potency of the therapeutic mechanism. The absence of significant off-target or systemic toxicity signals is a major de-risking factor for both the NTLA-2001 program and the underlying LNP-CRISPR platform. The observed AEs are largely predictable and consistent with the known profiles of LNP-formulated therapeutics, suggesting that the therapy is well-targeted and is not causing widespread, detectable cellular toxicity.

B. Analysis of Infusion-Related Reactions (IRRs) and Other Common Adverse Events

Infusion-related reactions (IRRs) have been the most common treatment-related adverse events observed in the program.[8] These reactions are a known class effect for LNP-based therapies. In the NTLA-2001 study, these events have been consistently described as predominantly mild to moderate in severity and transient in nature.[3] Importantly, all reported IRRs resolved without lasting clinical consequences (sequelae), and no patients have had to discontinue treatment due to an IRR.[8] This indicates that these reactions are clinically manageable, likely through standard measures such as premedication and careful control of the infusion rate.

C. Evaluation of Serious Adverse Events (SAEs) and Laboratory Findings

The program has maintained a very clean record with respect to serious adverse events. To date, only a single possibly related SAE has been reported. This was an event of Grade 3 vomiting in a patient in the 1.0 mg/kg dose cohort who had a relevant pre-existing medical condition, gastroparesis, which can predispose individuals to nausea and vomiting.[7] As a precautionary measure, the study protocol required an expansion of this dose cohort from three to six patients to further characterize safety. No additional related AEs of Grade 2 or higher were observed in the expanded cohort, providing confidence that this was likely an isolated event influenced by the patient's underlying condition.[10]

Crucially, there have been no clinically significant laboratory findings of concern, particularly with respect to liver function.[3] As the liver is the target organ for NTLA-2001, monitoring for hepatotoxicity is a key safety consideration. The absence of significant elevations in liver enzymes is a strong indicator of the therapy's safety and target specificity. While some transient, Grade 1 liver enzyme elevations were noted, these were not considered clinically significant and resolved spontaneously.[60]

While the short- and medium-term safety data are highly reassuring, the most significant unresolved question remains the long-term safety of permanent TTR protein knockout. TTR is a naturally occurring protein with known physiological roles, including the transport of vitamin A.[11] The immediate consequence of TTR reduction is a decrease in serum vitamin A levels, a known effect that is managed prophylactically with daily vitamin A supplementation for all trial participants.[61] However, the potential for unforeseen physiological consequences to emerge over decades due to the lifelong absence of TTR necessitates diligent and continuous monitoring. The clinical development program appropriately includes a separate, dedicated long-term follow-up study to monitor patients for any such effects, which will be essential for fully establishing the long-term safety of this innovative therapeutic approach.[3]

VII. Regulatory Trajectory and Market Positioning

NTLA-2001 is on an accelerated development pathway, supported by compelling clinical data and the recognition of its transformative potential by global regulatory agencies. Its unique profile as a "one-and-done" therapy positions it to disrupt the current market for ATTR amyloidosis treatments.

A. Key Regulatory Milestones: Orphan Drug and RMAT Designations

NTLA-2001 has received several key regulatory designations that validate its promise and are designed to expedite its development and review. These are not procedural formalities but are granted based on the strength of the preliminary clinical evidence and the therapy's potential to address a serious unmet medical need.

U.S. Food and Drug Administration (FDA):

  • Orphan Drug Designation: The FDA granted Orphan Drug Designation to NTLA-2001 for the treatment of ATTR amyloidosis.[63] This status is given to therapies for rare diseases affecting fewer than 200,000 people in the U.S. and provides significant development incentives, including tax credits, user fee waivers, and a period of seven years of market exclusivity upon approval.[63]
  • Regenerative Medicine Advanced Therapy (RMAT) Designation: In a strong signal of regulatory confidence, the FDA has granted RMAT designation to NTLA-2001 for both the ATTR-CM and ATTRv-PN indications.[66] The RMAT designation, established under the 21st Century Cures Act, is reserved for promising regenerative medicine therapies that have shown the potential to treat, modify, reverse, or cure a serious or life-threatening condition.[66] This designation confers significant benefits, including more frequent interactions with the FDA, eligibility for accelerated approval based on surrogate endpoints, and priority review of a future Biologics License Application (BLA).[67]
  • Investigational New Drug (IND) Clearance: The clearance of the IND application in October 2023 for the pivotal Phase 3 MAGNITUDE trial was a landmark event, officially marking NTLA-2001 as the first systemically administered in vivo CRISPR-based therapy to advance into late-stage clinical development.[4]

European Commission (EC):

  • Orphan Drug Designation: The EC also granted orphan drug designation for NTLA-2001 for the treatment of ATTR amyloidosis, providing similar development and market exclusivity incentives within the European Union.[72]

This collection of premier regulatory designations from the world's leading health authorities serves as a powerful external validation of the NTLA-2001 clinical data and its therapeutic potential. It materially accelerates the therapy's path to market and signals a high level of regulatory engagement and support.

B. Comparative Analysis: NTLA-2001 vs. TTR Stabilizers and RNA Silencers

NTLA-2001 is poised to enter a market with established, effective therapies. Its competitive positioning is defined by fundamental differences in its mechanism, administration, and ultimate therapeutic goal compared to the current standards of care.

Therapeutic ClassExample Drug(s)Mechanism of ActionAdministrationKey Efficacy Endpoint
Gene Knockout (CRISPR)NTLA-2001 (nex-z)Permanent inactivation of the TTR gene in hepatocytes, eliminating protein production at its source.3Single Intravenous Infusion 2>90% durable TTR reduction; potential to halt and reverse disease.6
Gene Silencers (siRNA/ASO)Patisiran, Vutrisiran, InotersenTransient degradation of TTR mRNA, reducing TTR protein synthesis.38Chronic infusions (every 3 weeks) or subcutaneous injections (weekly to quarterly).38~70-85% TTR reduction; slowing of neuropathy progression.38
TTR StabilizersTafamidis, AcoramidisBinds to the TTR tetramer, preventing its dissociation into pathogenic monomers.34Chronic daily oral administration.34No TTR reduction; slowing of cardiomyopathy progression, reduction in mortality/hospitalization.34

This comparison starkly illustrates the disruptive nature of NTLA-2001. Its primary competitive advantage lies not just in the depth of its TTR reduction but in its fundamental redefinition of the treatment paradigm. By shifting from a model of chronic, lifelong management to a single, potentially curative administration, NTLA-2001 offers a value proposition that transcends direct efficacy comparisons. The elimination of the immense burden associated with continuous therapy—including costs, adherence challenges, and the psychological impact—is a powerful differentiator that could drive rapid market adoption, assuming long-term safety is confirmed.

C. Future Outlook: Potential as a "One-and-Done" Curative Therapy

NTLA-2001 is positioned to be a transformative therapy that could dramatically reset the standard of care for ATTR amyloidosis.[2] If the ongoing Phase 3 trials successfully demonstrate that the profound and durable TTR reduction translates into a significant benefit on hard clinical outcomes like mortality and disease progression, NTLA-2001 could become the preferred treatment for newly diagnosed patients.

The "one-and-done" therapeutic model offers unparalleled advantages in patient convenience and adherence, freeing patients from the lifelong regimen of pills, injections, or infusions that characterize current treatments.[5] This unique profile is expected to command significant commercial interest, with market analysts forecasting that sales could reach $1.4 billion by 2029.[54] The success of NTLA-2001 would not only be a landmark achievement for patients with ATTR amyloidosis but would also serve as the ultimate validation for Intellia's systemic LNP-CRISPR platform, paving the way for the development of similar single-course, curative therapies for a wide range of other genetic diseases.

VIII. Conclusion and Expert Insights

A. Synthesis of Findings: The Transformative Potential of NTLA-2001

NTLA-2001 (nexiguran ziclumeran) has achieved a historic milestone in medicine, providing the first successful clinical proof-of-concept for a systemically delivered in vivo CRISPR/Cas9 gene-editing therapy. The data from its Phase 1 clinical program are unequivocal: a single intravenous infusion of NTLA-2001 can produce rapid, profound (>90%), and remarkably durable reductions in serum transthyretin, the protein at the root of ATTR amyloidosis. This potent pharmacodynamic effect, sustained for over two years in the longest-followed patients, is now being correlated with encouraging signs of clinical disease stabilization and improvement in both cardiac and neurological manifestations of the disease.

Combined with a favorable and manageable safety profile, these findings position NTLA-2001 as a highly promising, and potentially curative, treatment for all forms of ATTR amyloidosis, both hereditary and wild-type. The therapy's mechanism represents a paradigm shift, moving away from the chronic, lifelong management required by current standards of care toward a single-administration treatment designed to permanently resolve the underlying genetic cause. This "one-and-done" approach holds the potential to dramatically reduce treatment burden and transform the lives of patients with this devastating disease.

B. Critical Assessment of Unresolved Questions and Future Research Directions

Despite the profound promise of NTLA-2001, critical questions remain that must be addressed through continued rigorous clinical investigation.

  • Long-Term Safety: The most significant unresolved question is the long-term safety and physiological consequence of the lifelong absence of hepatic TTR production. While TTR's role in thyroxine transport is minor, its function as the primary transporter of retinol-binding protein is significant. Although managed with vitamin A supplementation, the full impact of its permanent absence over a human lifespan is unknown. The ongoing long-term follow-up studies are therefore of paramount importance to ensure no unforeseen health consequences emerge over decades.[3]
  • Disease Reversal: A pivotal area for future research is to determine whether the near-complete elimination of circulating TTR can lead to the regression of existing amyloid deposits and a true reversal of organ damage. While the current data show disease stabilization and some improvement, demonstrating the clearance of amyloid fibrils from tissues would represent the ultimate therapeutic achievement.[3] Advanced imaging and tissue biopsy studies will be essential to answer this question.
  • Broader Patient Populations: The initial clinical trials have focused on patients with a certain level of functional capacity (e.g., NYHA Class I-III). The efficacy and safety of NTLA-2001 in higher-risk patients, such as those with NYHA Class IV heart failure or severe renal impairment, who were excluded from the initial studies, remain to be determined.[3]

C. Recommendations for the Clinical and Research Communities

The NTLA-2001 program is a landmark in the history of genetic medicine. It has not only provided a potential cure for a devastating rare disease but has also successfully validated the systemic LNP-CRISPR platform as a viable therapeutic modality. The implications of this achievement extend far beyond ATTR amyloidosis, opening the door to the development of similar therapies for a wide array of other genetic diseases that originate in the liver.[3]

The clinical and research communities should monitor the progress of the pivotal Phase 3 MAGNITUDE and MAGNITUDE-2 trials with great attention. The outcomes of these studies will be instrumental in defining the future standard of care for ATTR amyloidosis and will have profound implications for the entire field of genomic medicine. Success in these trials would not only deliver a transformative new medicine to patients in need but would also herald the dawn of a new era where permanent, single-course genetic cures for inherited and age-related diseases become a clinical reality.

Works cited

  1. Intellia Announces Positive Clinical Proof-of-Concept Data for Redosing a CRISPR-Based Therapy with its Proprietary LNP-Based Delivery Platform, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-announces-positive-clinical-proof-concept-data-redosing
  2. Intellia Therapeutics Announces First Patient Dosed in the Phase 3 MAGNITUDE Study of NTLA-2001 as a Single-Dose CRISPR-Based Treatment for Transthyretin Amyloidosis with Cardiomyopathy, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-first-patient-dosed-phase-3
  3. Lessons from the first-in-human in vivo CRISPR/Cas9 editing of the ..., accessed October 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10624232/
  4. Intellia Therapeutics Announces FDA Clearance of Investigational New Drug (IND) Application to Initiate a Pivotal Phase 3 Trial of NTLA-2001 for the Treatment of Transthyretin (ATTR) Amyloidosis with Cardiomyopathy, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-fda-clearance-investigational-0
  5. NTLA-2001: opening a new era for gene therapy - PMC, accessed October 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11749809/
  6. In vivo CRISPR/Cas9 Editing of the TTR Gene by NTLA-2001 in Patients with Transthyretin Amyloidosis - Intellia Therapeutics, accessed October 4, 2025, https://www.intelliatx.com/wp-content/uploads/PNS-Oral-Pres_Gillmore_062621.pdf
  7. Intellia and Regeneron Present Updated Interim Data from Phase 1 Study of CRISPR-based NTLA-2001 for the Treatment of Transthyretin (ATTR) Amyloidosis Demonstrating that Deep Serum TTR Reductions Remained Durable After a Single Dose, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-and-regeneron-present-updated-interim-data-phase-1
  8. Intellia Announces First Clinical Evidence from Ongoing Phase 1 ..., accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-announces-first-clinical-evidence-ongoing-phase-1-study
  9. Intellia Announces Positive Two-Year Follow-Up Data from Ongoing Phase 1 Study of Nexiguran Ziclumeran (nex-z), in Patients with Hereditary Transthyretin (ATTR) Amyloidosis with Polyneuropathy at Peripheral Nerve Society Annual Meeting, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-announces-positive-two-year-follow-data-ongoing-phase-1
  10. Intellia and Regeneron Announce Updated Phase 1 Data Demonstrating a Single Dose of NTLA-2001, an Investigational CRISPR Therapy for Transthyretin (ATTR) Amyloidosis, Resulted in Rapid, Deep and Sustained Reduction in Disease-Causing Protein, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-and-regeneron-announce-updated-phase-1-data
  11. Transthyretin-Related Amyloidosis: Background, Pathophysiology, Epidemiology, accessed October 4, 2025, https://emedicine.medscape.com/article/335301-overview
  12. Transthyretin amyloidosis: MedlinePlus Genetics, accessed October 4, 2025, https://medlineplus.gov/genetics/condition/transthyretin-amyloidosis/
  13. Mechanism of Action and Clinical Application of Tafamidis in Hereditary Transthyretin Amyloidosis - PMC - PubMed Central, accessed October 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4919130/
  14. Hereditary ATTR Amyloidosis, accessed October 4, 2025, https://arci.org/about-amyloidosis/hereditary-attr-amyloidosis/
  15. ATTR Pathophysiology | ATTR HCP - ATTR With Cardiomyopathy Homepage, accessed October 4, 2025, https://www.attr-amyloidosis.com/disease-overview/pathophysiology
  16. Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade | PNAS, accessed October 4, 2025, https://www.pnas.org/doi/10.1073/pnas.1121005109
  17. HEREDITARY TRANSTHYRETIN AMYLOIDOSIS, accessed October 4, 2025, https://arci.org/wp-content/uploads/2021/03/Disease-Overview_hATTR.pdf
  18. Transthyretin Amyloidosis: The Importance and Need for Early Diagnosis - AstraZeneca US, accessed October 4, 2025, https://www.astrazeneca-us.com/media/astrazeneca-us-blog/2024/transthyretin-amyloidosis-the-importance-and-need-for-early-diagnosis.html
  19. Transthyretin Amyloidosis (ATTR) - Pfizer, accessed October 4, 2025, https://www.pfizer.com/disease-and-conditions/attr-amyloidosis
  20. Lessons from the first-in-human in vivo CRISPR/Cas9 editing of the TTR gene by NTLA-2001 trial in patients with transthyretin amyloidosis with cardiomyopathy - PubMed, accessed October 4, 2025, https://pubmed.ncbi.nlm.nih.gov/37928601/
  21. Diagnosis and Treatment of Hereditary Transthyretin Amyloidosis (hATTR) Polyneuropathy: Current Perspectives on Improving Patient Care - PubMed Central, accessed October 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7041433/
  22. Transthyretin Familal Amyloid Neuropathy - - Practical Neurology, accessed October 4, 2025, https://practicalneurology.com/diseases-diagnoses/neuromuscular/transthyretin-familal-amyloid-neuropathy/31688/
  23. Wild-Type Amyloidosis, accessed October 4, 2025, https://arci.org/about-amyloidosis/wild-type-amyloidosis/
  24. What Is Wild-Type ATTR Amyloidosis? - Everyday Health, accessed October 4, 2025, https://www.everydayhealth.com/amyloidosis/wild-type-attr/
  25. Wild-Type (Senile) ATTR Amyloidosis - Stanford Health Care, accessed October 4, 2025, https://stanfordhealthcare.org/medical-conditions/blood-heart-circulation/amyloidosis/types/wild-type-senile-attr-amyloidosis.html
  26. Transthyretin amyloid cardiomyopathy (ATTR-CM) - University Hospitals, accessed October 4, 2025, https://www.uhhospitals.org/services/heart-and-vascular-services/conditions-and-treatments/cardiomyopathy/transthyretin-amyloid-cardiomyopathy
  27. Transthyretin Amyloid Cardiomyopathy (ATTR-CM) - American Heart Association, accessed October 4, 2025, https://www.heart.org/en/health-topics/cardiomyopathy/what-is-cardiomyopathy-in-adults/transthyretin-amyloid-cardiomyopathy-attr-cm
  28. Cardiac Amyloidosis: Evolving Diagnosis and Management: A Scientific Statement From the American Heart Association | Circulation, accessed October 4, 2025, https://www.ahajournals.org/doi/10.1161/CIR.0000000000000792
  29. Transthyretin Amyloid Cardiomyopathy: JACC State-of-the-Art Review - PMC, accessed October 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6724183/
  30. What Is the Difference Between Hereditary vs. Wild Type ATTR-CM? - Health Central, accessed October 4, 2025, https://www.healthcentral.com/slideshow/difference-between-hereditary-vs-wild-type-attr-cm
  31. Hereditary Transthyretin Amyloidosis (hATTR) With Polyneuropathy - WebMD, accessed October 4, 2025, https://www.webmd.com/brain/hereditary-transthyretin-amyloidosis-polyneuropathy
  32. Current Landscape of Therapies for Transthyretin Amyloid Cardiomyopathy - JACC Journals, accessed October 4, 2025, https://www.jacc.org/doi/10.1016/j.jchf.2025.03.017
  33. Transthyretin Amyloidosis (ATTR-CM): Types, Causes, Treatment - Cleveland Clinic, accessed October 4, 2025, https://my.clevelandclinic.org/health/diseases/17855-amyloidosis-attr
  34. Tafamidis - StatPearls - NCBI Bookshelf, accessed October 4, 2025, https://www.ncbi.nlm.nih.gov/books/NBK574508/
  35. Current and emerging treatment options for transthyretin amyloid cardiomyopathy - Heart, accessed October 4, 2025, https://heart.bmj.com/content/early/2025/05/26/heartjnl-2024-325184
  36. Transthyretin-Related Amyloidosis Treatment & Management - Medscape Reference, accessed October 4, 2025, https://emedicine.medscape.com/article/335301-treatment
  37. TTR Stabilization | How VYNDAMAX (tafamidis) Works | Safety Info, accessed October 4, 2025, https://vyndamax.pfizerpro.com/about-vyndamax/how-vyndamax-works
  38. Patisiran - StatPearls - NCBI Bookshelf, accessed October 4, 2025, https://www.ncbi.nlm.nih.gov/books/NBK589768/
  39. Vutrisiran - Wikipedia, accessed October 4, 2025, https://en.wikipedia.org/wiki/Vutrisiran
  40. Inotersen - StatPearls - NCBI Bookshelf, accessed October 4, 2025, https://www.ncbi.nlm.nih.gov/books/NBK578206/
  41. FDA Approves Amvuttra™ (vutrisiran) for ATTR-CM: More Options for Patients, accessed October 4, 2025, https://arci.org/amvuttra-attr-cm-approval/
  42. 211172Orig1s000 - accessdata.fda.gov, accessed October 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/211172Orig1s000SumR.pdf
  43. Vutrisiran (Amvuttra) - NCBI Bookshelf, accessed October 4, 2025, https://www.ncbi.nlm.nih.gov/books/NBK602494/
  44. MAGNITUDE: A Phase 3 Multinational Multicenter Randomized Double-Blind Placebo-Controlled Study to Evaluate the Efficacy and Safety of NTLA-2001 in Participants with Transthyretin Amyloidosis with Cardiomyopathy (ATTR-CM) - Clinical Trials, accessed October 4, 2025, https://clinicaltrials.med.nyu.edu/clinicaltrial/2418/magnitude-phase-3-multinational/
  45. NTLA-2001: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 4, 2025, https://go.drugbank.com/drugs/DB18525
  46. Intellia Therapeutics Announces Second Quarter 2024 Financial Results and Highlights Recent Company Progress, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-second-quarter-2024-financial
  47. Intellia opts out of hemophilia B gene editing work with Regeneron - Fierce Biotech, accessed October 4, 2025, https://www.fiercebiotech.com/biotech/intellia-opts-out-hemophilia-gene-editing-work-regeneron
  48. Intellia and Regeneron Announce Landmark Clinical Data Showing Deep Reduction in Disease-Causing Protein After Single Infusion of NTLA-2001, an Investigational CRISPR Therapy for Transthyretin (ATTR) Amyloidosis, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-and-regeneron-announce-landmark-clinical-data-showing
  49. NCT04601051 | Study to Evaluate Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of NTLA-2001 in Patients With Hereditary Transthyretin Amyloidosis With Polyneuropathy (ATTRv-PN) and Patients With Transthyretin Amyloidosis-Related Cardiomyopathy (ATTR-CM) | ClinicalTrials.gov, accessed October 4, 2025, https://clinicaltrials.gov/study/NCT04601051
  50. Intellia and Regeneron Announce Initial Data from the Cardiomyopathy Arm of Ongoing Phase 1 Study of NTLA-2001, an Investigational CRISPR Therapy for the Treatment of Transthyretin (ATTR) Amyloidosis, accessed October 4, 2025, https://amyloidosis.org/intellia-and-regeneron-announce-initial-data-from-the-cardiomyopathy-arm-of-ongoing-phase-1-study-of-ntla-2001-an-investigational-crispr-therapy-for-the-treatment-of-transthyretin-attr-amyloidosis/
  51. Intellia Therapeutics Doses First Patient in Landmark CRISPR/Cas9 Clinical Trial of NTLA-2001 for the Treatment of Transthyretin Amyloidosis, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-doses-first-patient-landmark-crisprcas9
  52. Intellia and Regeneron Announce Landmark Clinical Data Showing Deep Reduction in Disease-Causing Protein After Single Infusion of NTLA-2001, an Investigational CRISPR Therapy for Transthyretin (ATTR) Amyloidosis - SEC.gov, accessed October 4, 2025, https://www.sec.gov/Archives/edgar/data/1652130/000119312521201044/d197054dex991.htm
  53. NCT06128629 | MAGNITUDE: A Phase 3 Study of NTLA-2001 in Participants With Transthyretin Amyloidosis With Cardiomyopathy (ATTR-CM) | ClinicalTrials.gov, accessed October 4, 2025, https://clinicaltrials.gov/study/NCT06128629
  54. Intellia Therapeutics doses first patient in pivotal in vivo CRISPR trial, accessed October 4, 2025, https://www.clinicaltrialsarena.com/news/intellia-therapeutics-crispr-first-patient-pivotal/
  55. Clinical Trials - Intellia Therapeutics, accessed October 4, 2025, https://www.intelliatx.com/pipeline/clinical-trials/
  56. Intellia Therapeutics Announces First Patient Dosed in the MAGNITUDE-2 Phase 3 Study of Nexiguran Ziclumeran (nex-z), a One-Time Gene Editing-Based Treatment for Transthyretin (ATTR) Amyloidosis with Polyneuropathy, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-first-patient-dosed-magnitude-2
  57. Intellia Therapeutics Announces Anticipated 2025 Milestones and Strategic Reorganization to Prioritize the Advancement of its Late-Stage Programs, NTLA-2002 and Nexiguran Ziclumeran (nex-z), accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-anticipated-2025-milestones-and
  58. Intellia's gene editing therapy shows early potential in rare heart condition, accessed October 4, 2025, https://www.clinicaltrialsarena.com/news/intellias-gene-editing-therapy-shows-early-potential-in-rare-heart-condition/
  59. Intellia's Phase 3 Clinical Trial for CRISPR-Based Gene Therapy Enrolls First Patient, accessed October 4, 2025, https://www.cgtlive.com/view/intellia-phase-3-clinical-trial-crispr-based-gene-therapy-enrolls-first-patient
  60. NTLA-2001 for ATTR Amyloidosis: Interim Clinical Results from ..., accessed October 4, 2025, https://ir.intelliatx.com/static-files/514441ee-8fbc-45a4-ac36-3c9edf3798dc
  61. MAGNITUDE: A Phase 3 Study of NTLA-2001 in Participants With Transthyretin Amyloidosis With Cardiomyopathy (ATTR-CM) - Stanford Health Care, accessed October 4, 2025, https://stanfordhealthcare.org/trials/m/NCT06128629.html
  62. MAGNITUDE: A Phase 3 Study of NTLA-2001 in Participants With Transthyretin Amyloidosis With Cardiomyopathy (ATTR-CM) | Clinical Research Trial Listing - CenterWatch, accessed October 4, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT06128629/magnitude-a-phase-3-study-of-ntla-2001-in-participants-with-transthyretin-amyloidosis-with-cardiomyopathy-attr-cm
  63. Intellia Therapeutics Receives U.S. FDA Orphan Drug Designation for NTLA-2001, an Investigational CRISPR Therapy for the Treatment of Transthyretin (ATTR) Amyloidosis, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-receives-us-fda-orphan-drug-designation
  64. Intellia's Milestone In Vivo CRISPR Therapy Granted FDA Orphan Drug Designation, accessed October 4, 2025, https://www.biospace.com/intellia-s-milestone-in-vivo-crispr-therapy-granted-fda-orphan-drug-designation
  65. Intellia Therapeutics Receives U.S. FDA Orphan Drug Designation for NTLA-5001 for the Treatment of Acute Myeloid Leukemia, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-receives-us-fda-orphan-drug-designation-0
  66. Intellia Therapeutics Announces FDA Regenerative Medicine Advanced Therapy (RMAT) Designation Granted to Nexiguran Ziclumeran (nex-z) for the Treatment of Transthyretin (ATTR) Amyloidosis with Cardiomyopathy, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-fda-regenerative-medicine-1
  67. Intellia Therapeutics Announces FDA Regenerative Medicine Advanced, accessed October 4, 2025, https://amyloidosis.org/intellia-therapeutics-announces-fda-regenerative-medicine-advanced-therapy-rmat-designation-granted-to-nexiguran-ziclumeran-nex-z-for-the-treatment-of-transthyretin-attr-amyloidosis-with-cardiom/
  68. Updated FDA granted RMAT List (131) - The Niche - Stem Cell Blog, accessed October 4, 2025, https://ipscell.com/rmat-list/
  69. Regenerative Medicine Advanced Therapy Designation - FDA, accessed October 4, 2025, https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/regenerative-medicine-advanced-therapy-designation
  70. FDA Designates Intellia's Nexiguran Ziclumeran (nex-z) as a Regenerative Medicine Advanced Therapy | Goodwin - JD Supra, accessed October 4, 2025, https://www.jdsupra.com/legalnews/fda-designates-intellia-s-nexiguran-3763240/
  71. Intellia Therapeutics Announces Third Quarter 2023 Financial Results and Highlights Recent Company Progress - SEC.gov, accessed October 4, 2025, https://www.sec.gov/Archives/edgar/data/1652130/000095017023061539/ntla-ex99_1.htm
  72. Intellia Therapeutics Announces FDA Regenerative Medicine Advanced Therapy (RMAT) Designation Granted to Nexiguran Ziclumeran (nex-z) for the Treatment of Hereditary Transthyretin (ATTR) Amyloidosis with Polyneuropathy, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-announces-fda-regenerative-medicine-0
  73. Intellia Therapeutics' Investigational CRISPR Treatment NTLA-2001 Receives European Union Orphan Drug Designation for ATTR Amyloidosis, accessed October 4, 2025, https://ir.intelliatx.com/news-releases/news-release-details/intellia-therapeutics-investigational-crispr-treatment-ntla-2001

Published at: October 4, 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.