Nexiguran Ziclumeran Gene Editing in Hereditary ATTR with Polyneuropathy
Click here to hear in youtube: https://youtu.be/xo5UM5ey2f4
Reference: New England Journal of Medicine. 2025; DOI: 10.1056/NEJMoa2510209
Executive Summary
The New England Journal of Medicine (NEJM) has published results of the pivotal trial evaluating nexiguran ziclumeran, an in vivo CRISPR-Cas9–based therapy targeting transthyretin (TTR) for hereditary transthyretin amyloidosis with polyneuropathy (ATTRv-PN). The therapy, delivered via lipid nanoparticles, aims at permanent silencing of hepatic TTR production through gene editing.
The trial demonstrated sustained and clinically meaningful reductions in serum TTR levels (>90% median knockdown) with accompanying stabilization or improvement of neuropathy scores. Safety data were generally favorable, though mild transient elevations of liver enzymes were observed. Importantly, no evidence of off-target editing or long-term genotoxicity has yet emerged, but follow-up remains short relative to the lifelong implications of genome editing.
This publication marks a watershed: from RNA interference (patisiran, vutrisiran) and antisense oligonucleotides (inotersen, eplontersen) toward one-time, potentially curative gene editing for ATTRv. The broader implications extend beyond amyloidosis, testing the limits of in vivo CRISPR translation into chronic disease therapeutics.
Five Laws of Epistemic Integrity
1. Truthfulness of Information (Factual Rigor)
Data confirm deep and sustained TTR silencing, consistent with the proposed mechanism. Neuropathy measures show stabilization, aligning with pharmacodynamic expectations. However, trial duration (~12–18 months) is insufficient to claim “cure.”
Verdict: High Integrity
2. Source Referencing (Transparency of Methods)
The NEJM article discloses design, randomization, endpoints, and statistical analysis in detail. Methodological transparency is reinforced by supplementary data (CRISPR editing specificity, sequencing analyses).
Verdict: High Integrity
3. Reliability & Accuracy (Reproducibility)
The study is multicenter and international, but sample size remains modest (<100 patients). External validity beyond ATTRv-PN (to wild-type ATTR, cardiomyopathy phenotypes) is untested. Reproducibility across ethnic, metabolic, and genetic contexts is uncertain.
Verdict: Moderate Integrity
4. Contextual Judgment (Limitations & Scope)
Authors acknowledge limitations: follow-up is too short, rare off-target events may take years to manifest, and the long-term consequences of permanent TTR suppression are unknown (e.g., TTR has physiological roles in vitamin A and thyroxine transport).
Verdict: High Integrity
5. Inference Traceability (Policy & Implementation)
The leap from interim trial success to global adoption faces gaps: long-term safety, manufacturing scale, pricing, and health system readiness for gene editing. Policy implications (coverage, regulatory frameworks, equity of access) are not yet mapped.
Verdict: Moderate Integrity
BBIU Opinion – Gaps Between Protocol and Clinical Reality
The NCT04601051 trial (NTLA-2001) was groundbreaking as the first in-human demonstration of in vivo CRISPR editing. Yet, as expected for an early-phase, dose-escalation design, the protocol emphasized immediate safety and proof of pharmacodynamic effect, while leaving critical questions on SAE attribution unresolved.
What the protocol did include
Standard safety monitoring: AE/SAE collection, hepatic labs, treatment-emergent adverse events.
Conventional causality assessment (related / possibly / unlikely / not related).
Dose escalation with tight observation of safety labs and TTR knockdown.
What the protocol did not pre-specify
No formal framework to differentiate SAEs caused by ATTRv progression from those induced by Cas9/LNP delivery.
No stratification or adjustment by age or TTR burden (disease duration, cardio-neuro phenotype).
No systematic plan for anti-Cas9 immunogenicity profiling or robust off-target detection.
Why this matters
SAE attribution is left largely to individual investigator judgment, limiting epistemic traceability.
In older patients, where cumulative exposure to TTR is higher, the probability of disease-driven SAEs rises—but the protocol did not account for this prospectively.
The structure of dose escalation carries interpretive weight: clustering of SAEs at higher doses would implicate Cas9/LNP, while random distribution across doses points toward ATTRv progression. Yet the protocol did not define rules to interrogate this distinction.
The absence of a placebo arm is ethically justified: in ATTRv-PN, with no curative options, placebo would offer no clinical benefit. But this also reinforces that the trial’s purpose was less about comparative efficacy and more about validating safety and biological activity.
Most importantly, this trial is as much about proving systemic CRISPR delivery as it is about treating ATTRv-PN. Participants occupy a dual role: patients seeking relief and symbolic pioneers advancing a platform. Their long-term outcomes will matter disproportionately.
Long-Term Surveillance and Equity
The participants’ importance does not end with the trial report. They are the first reference cohort for systemic gene editing in humans. Multi-year surveillance is essential: durability of TTR suppression, delayed immunogenicity, or rare off-target events may emerge only after a decade. Equally, policy makers must address equity: if costs approach or exceed USD 1 million per treatment, will access be limited to wealthy health systems, or can gene editing be democratized? Without answers, clinical success may widen global disparities rather than close them.
Annex – Understanding Cas9
Cas9 is a protein that originally comes from bacteria, where it works like part of a defense system against viruses. Scientists discovered that this natural tool can be repurposed in medicine to edit human genes. Cas9 is often described as “molecular scissors,” but its function is more precise: it is programmable and can be directed to almost any chosen spot in the genome.
How Cas9 Finds Its Target
Cas9 follows two guiding rules:
An RNA guide – designed in the laboratory, it acts like a GPS that directs Cas9 to the right address in the genome.
A PAM signal – a short DNA tag (“NGG” for SpCas9) that acts like an entry ticket. Without this ticket, Cas9 ignores the site even if the RNA map points there.
Only when Cas9 finds both the entry ticket and a perfect match to its GPS map does it cut.
How Cas9 Cuts
Once bound, Cas9 opens the DNA and cuts both strands—imagine unzipping a zipper and then slicing through both sides. The cell then repairs the break: sometimes sloppily (disabling a gene), sometimes with new instructions provided by scientists.
Why Cas9 Is Different
Traditional enzymes are like padlock keys: each fits one exact lock. Cas9 is more like a universal key that can be reprogrammed by changing the RNA guide, making it far more versatile.
Accuracy and Challenges
Cas9 is powerful but not flawless:
A poorly designed GPS (RNA guide) can misdirect it.
Different Cas9 “models” have different accuracy (some engineered for fewer mistakes).
DNA wrapped tightly in chromatin is harder to reach.
This is why scientists spend as much effort on off-target detection as on the main experiment.
Making Cas9 Safe in Humans
For clinical use, Cas9 is not injected as a raw protein. Instead, the instructions to make it are packed into lipid nanoparticles (tiny fat bubbles). These are naturally decorated with ApoE proteins in the bloodstream, guiding them to the liver, where most TTR is made. Liver cells briefly produce Cas9, cut the TTR gene, and then degrade it. Exposure is temporary—days, not months—reducing long-term risks.
Why This Matters
Cas9 is more than “scissors.” It is a programmable editing system that combines:
GPS navigation (RNA guide),
an entry ticket (PAM), and
a cutting tool (the enzyme itself).
The same Cas9 can be reused for many diseases simply by swapping the GPS map. But precision matters: every cut is permanent, and every mistake leaves a lifelong trace.
This is why the first patients who received Cas9 therapy are not only medical subjects but historical participants. Their outcomes will decide whether genome editing transitions from promise to practice in medicine.