Fact checked byRichard Smith

Read more

November 05, 2022
2 min read
Save

CRISPR-based genome editing therapy may help in ATTR amyloidosis with cardiomyopathy

Fact checked byRichard Smith
You've successfully added to your alerts. You will receive an email when new content is published.

Click Here to Manage Email Alerts

We were unable to process your request. Please try again later. If you continue to have this issue please contact customerservice@slackinc.com.

CHICAGO — A novel CRISPR-based in vivo gene editing therapy reduced transthyretin levels in patients with hereditary transthyretin amyloidosis with cardiomyopathy, researchers reported at the American Heart Association Scientific Sessions.

Perspective from Alex Reyentovich, MD

Transthyretin amyloid cardiomyopathy (ATTR-CM) is a “progressive and fatal disease,” during which amyloid deposits cause impaired systolic and diastolic function and conduction disorders, Julian D. Gillmore, MBBS, MD, PhD, director of the Centre for Amyloidosis at University College London, said during a press conference. He said ATTR-CM is usually fatal within 3 to 10 years if untreated.

genome_sequencing_adobe_resized
A novel CRISPR-based in vivo gene editing therapy reduced transthyretin levels in patients with hereditary transthyretin amyloidosis with cardiomyopathy.
Source: Adobe Stock

“There remains a major unmet medical need in ATTR cardiomyopathy,” he said. “Current treatments only slow disease progression and require lifelong administration, and due to cost, access to treatment is limited.”

In a first-in-human study, the researchers evaluated NTLA-2001 (Intellia/Regeneron), a novel CRISPR/Cas9-based in vivo gene editing therapy. The primary objectives were safety, tolerability, pharmacokinetics, pharmacodynamics and effect on serum transthyretin (TTR) levels. The secondary objectives were efficacy on clinical measures including cardiac imaging, biomarkers, cardiopulmonary exercise testing and 6-minute walk distance.

The study included 12 patients (median age, 75 years; 100% men) with hereditary or wild-type ATTR-CM and NYHA class I to III HF who were administered a single dose of NTLA-2001 via IV infusion.

“This is a cohort in whom reduction in serum TTR protein concentration has recently been shown to be of clinical benefit,” Gillmore said. “The anticipation is that greater reduction will translate into greater clinical benefit. The hypothesis of knockout of the TTR gene to achieve deep sustained TTR reduction is an attractive one.”

Six patients with NYHA class III received a dose of 0.7 mg/kg. Three patients with NYHA class I or II received a dose of 0.7 mg/kg and three with NYHA class I or II received a dose of 1 mg/kg.

Three patients reported no adverse events and eight reported mild or moderate adverse events, according to Gillmore. One patient with NYHA class III who received a dose of 0.7 mg/kg had a grade 3 infusion-related reaction, which resolved without any clinical sequelae, Gillmore said.

There were no clinically significant laboratory findings, Gillmore said.

Serum TTR declined by 93% at 6 months in patients with NYHA class I or II who received a dose of 0.7 mg/kg, by 92% at 4 months in patients NYHA class I or II who received a dose of 1 mg/kg and by 94% in patients with NYHA class III who received a dose of 0.7 mg/kg, Gillmore said. All patients achieved a serum TTR reduction of at least 90% by 28 days.

The results are similar to those of patients with polyneuropathy who received NTLA-2001 in a separate arm of the trial, Gillmore said.

“Deep, consistent and durable TTR reductions were achieved at both the 0.7 and 1 mg/kg doses, with a TTR reduction of greater than 90% achieved at both doses by day 28 and out to the latest follow-up between 4 and 6 months,” Gillmore said. “NTLA-2001 was generally well tolerated at both doses and had similar results in patients with NYHA class I/II and NYHA class III. These data demonstrate the promise of CRISPR and Cas9-based in vivo gene editing in humans.”