Rapirosiran: A Promising RNA Interference Therapy Targeting HSD17B13 for Metabolic Dysfunction-Associated Steatohepatitis

Rapirosiran: A Promising RNA Interference Therapy Targeting HSD17B13 for Metabolic Dysfunction-Associated Steatohepatitis

Highlight

  • Rapirosiran is an investigational small-interfering RNA targeting liver-expressed HSD17B13 mRNA to treat MASH.
  • Phase I trial showed rapirosiran was well tolerated with no serious treatment-related adverse events.
  • Rapirosiran induced a dose-dependent reduction in hepatic HSD17B13 mRNA, reaching a median reduction of 78% at six months with the highest dose.
  • This novel RNA interference therapeutic represents a potential new treatment avenue for MASH, which currently has limited pharmacological options.

Study Background and Disease Burden

Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as nonalcoholic steatohepatitis (NASH), is a progressive liver disease characterized by hepatic steatosis, inflammation, and varying degrees of fibrosis. It poses a significant health burden globally due to its high prevalence linked to obesity and metabolic syndrome. MASH is a leading cause of liver-related morbidity and mortality, including cirrhosis and hepatocellular carcinoma, and currently lacks widely approved pharmacological therapies, with lifestyle modification remaining the mainstay.

Genetic factors modulate the risk and progression of chronic liver diseases. Genome-wide association studies identified loss-of-function variants in the hydroxysteroid 17-beta dehydrogenase 13 gene (HSD17B13) as protective against chronic liver disease, including MASH. HSD17B13 encodes a liver-specific enzyme involved in lipid metabolism and retinol dehydrogenase activities, implicating it in the pathogenesis of fatty liver disease. Therapeutically targeting HSD17B13 by reducing its hepatic expression offers a novel strategy for MASH treatment.

Study Design

The ALN-HSD-001 study was a randomized, double-blind, placebo-controlled, multicenter phase I trial designed to evaluate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of rapirosiran, an N-acetylgalactosamine-conjugated small-interfering RNA (siRNA) targeting HSD17B13 mRNA.

The study was conducted in two parts:

Part A involved 58 healthy adult volunteers who received single ascending subcutaneous doses of rapirosiran or placebo to characterize safety and PK profile.

Part B enrolled 46 adults diagnosed with MASH. Participants received two doses of rapirosiran or placebo administered 12 weeks apart. All MASH patients underwent liver biopsies at screening and post-randomization for quantitative measurement of hepatic HSD17B13 mRNA expression, allowing direct assessment of target engagement.

The primary endpoint was the frequency and severity of adverse events (AEs). Secondary endpoints included plasma and urinary pharmacokinetics and the change from baseline in liver HSD17B13 mRNA expression.

Key Findings

Safety and Tolerability:

– In Part A (healthy adults), injection-site reactions were the only adverse events occurring in ≥10% of rapirosiran-treated individuals (11%). These reactions were mild and transient.
– No serious treatment-related adverse events (SAEs) occurred during the study.
– In Part B (MASH patients), the only AE reported in ≥10% of rapirosiran-treated subjects was COVID-19 infection (14%), which was unrelated to the study drug.
– Importantly, no drug-induced liver injury was detected in either part.

Pharmacokinetics:

– Rapirosiran plasma concentrations declined rapidly within 24 hours post-dose, consistent with expected siRNA pharmacokinetics.
– Renal excretion accounted for 17%-37% of rapirosiran clearance across doses.

Pharmacodynamics and Efficacy:

– In MASH patients, rapirosiran demonstrated a clear dose-dependent reduction in liver HSD17B13 mRNA expression.
– The highest dose group (400 mg) exhibited a median reduction of 78% in liver HSD17B13 mRNA expression at six months post-treatment initiation.
– This robust hepatic target engagement supports the hypothesized mechanism of action.

Expert Commentary

This first-in-human trial of rapirosiran highlights the therapeutic potential of RNA interference in modulating key pathogenic drivers of MASH. The favorable safety profile is reassuring, especially in a population with underlying liver pathology where drug-induced liver injury is a concern. The substantial reduction of hepatic HSD17B13 mRNA validates effective delivery of the siRNA to the liver and successful gene silencing.

Loss-of-function genetic variants in HSD17B13 have been associated with protection from progressing chronic liver disease, suggesting reduced enzyme activity as beneficial. Rapirosiran’s pharmacodynamics reflect this protective mechanism by directly lowering hepatic HSD17B13 expression. This gene-targeted therapeutic approach is innovative compared to prior metabolic or inflammatory modulators tested in MASH.

Limitations of this phase I study include the small sample sizes and short-term follow-up, which preclude assessment of long-term clinical outcomes such as liver histology improvement or fibrosis regression. While liver biopsy confirmation of mRNA knockdown is a strength, clinical relevance will ultimately depend on demonstration of meaningful disease modification in future larger, longer studies.

Conclusion

Rapirosiran, a novel N-acetylgalactosamine-conjugated siRNA therapeutic targeting HSD17B13, demonstrates encouraging safety, tolerability, and pharmacokinetics in healthy and MASH populations. Its robust, dose-dependent hepatic mRNA knockdown confirms targeted delivery and gene silencing. This study lays the groundwork for continued clinical development of rapirosiran aiming to address the significant unmet need for effective pharmacotherapy in MASH, a disease with limited approved treatment options.

Ongoing and future studies should evaluate the clinical efficacy, optimal dosing regimen, and long-term safety to determine whether modulation of HSD17B13 can alter disease progression and improve patient outcomes in MASH.

References

Sanyal AJ, Taubel J, Badri P, Bond S, Makarova N, Zhao W, Duggal S, Kajbaf F, Olenchock BA, Gansner JM. Phase I randomized double-blind study of an RNA interference therapeutic targeting HSD17B13 for metabolic dysfunction-associated steatohepatitis. J Hepatol. 2025 Oct;83(4):838-848. doi: 10.1016/j.jhep.2025.05.031. Epub 2025 Jun 27. PMID: 40581300.

Younossi ZM, et al. Global epidemiology of nonalcoholic fatty liver disease—Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016 Jul;64(1):73-84.

Manns MP, et al. Non-alcoholic steatohepatitis: a global health problem. J Hepatol. 2016 Sep; 65(3): 479-491.

Musso G, et al. Genetic and epigenetic factors in the pathogenesis of metabolic-associated fatty liver disease. Nat Rev Gastroenterol Hepatol. 2023 Jun;20(6):402-414.

Emerging Therapies for NASH: Mechanisms and Clinical Trials. Lancet Gastroenterol Hepatol. 2023 Jan;8(1):27-38.

Rapirosiran: A Promising RNAi Therapy Targeting HSD17B13 in Metabolic Dysfunction-Associated Steatohepatitis

Rapirosiran: A Promising RNAi Therapy Targeting HSD17B13 in Metabolic Dysfunction-Associated Steatohepatitis

Highlight

  • Rapirosiran is a novel, N-acetylgalactosamine (GalNAc)-conjugated small interfering RNA (siRNA) targeting the liver-expressed HSD17B13 gene, implicated in chronic liver disease.
  • In a randomized, double-blind, placebo-controlled Phase I trial (ALN-HSD-001), rapirosiran showed a favorable safety and tolerability profile in both healthy adults and adults with metabolic dysfunction-associated steatohepatitis (MASH).
  • Pharmacodynamic assessment revealed a robust, dose-dependent reduction of up to 78% in liver HSD17B13 mRNA expression after two doses over six months in MASH patients.
  • These data support further clinical development of rapirosiran as a potential therapeutic option for MASH, addressing a significant unmet medical need.

Study Background and Disease Burden

Metabolic dysfunction-associated steatohepatitis (MASH), previously termed non-alcoholic steatohepatitis (NASH), represents a progressive form of chronic liver disease characterized by hepatic steatosis, inflammation, and fibrotic changes. MASH is strongly associated with metabolic syndrome components, including obesity, type 2 diabetes mellitus, and dyslipidemia, and is projected to become the leading indication for liver transplantation globally. Despite its high prevalence and substantial morbidity and mortality, there remains only a single approved pharmacological treatment option, underscoring a critical unmet need for effective therapies.

Genome-wide association studies have implicated loss-of-function variants in the hydroxysteroid 17-beta dehydrogenase 13 gene (HSD17B13) as being protective against chronic liver disease, including fibrosis and hepatocellular carcinoma. HSD17B13 encodes a liver-specific lipid droplet-associated enzyme believed to modulate lipid metabolism and inflammatory pathways relevant to MASH pathogenesis. These human genetic findings provide a compelling rationale for the therapeutic targeting of HSD17B13 to ameliorate disease progression.

Rapirosiran is an investigational RNA interference (RNAi) therapeutic designed to selectively degrade HSD17B13 mRNA in hepatocytes. By reducing hepatic expression of this pathogenic gene, rapirosiran aims to mitigate liver injury and fibrosis in affected individuals.

Fig. 1

Fig. 1 Study design.

Study Design

The ALN-HSD-001 trial was a two-part, randomized, double-blind, placebo-controlled, multicenter Phase I study.

-part A enrolled 58 healthy adults who received single ascending subcutaneous doses of rapirosiran or placebo to characterize safety, tolerability, and pharmacokinetics (PK).

-part B enrolled 46 adults with biopsy-confirmed MASH who received two subcutaneous doses of rapirosiran or placebo 12 weeks apart. Liver biopsies were performed at screening and after treatment to quantify hepatic HSD17B13 mRNA levels.

The primary endpoint in both parts was the frequency and severity of adverse events (AEs), focusing on safety and tolerability. Secondary endpoints included PK profiling of plasma and urine rapirosiran concentrations and changes from baseline in liver HSD17B13 mRNA expression.

Fig. 2

Fig. 2 Disposition of healthy participants (Part A) and patients with metabolic dysfunction-associated steatohepatitis (MASH; Part B).

Key Findings

Safety and Tolerability
In Part A (healthy adults), rapirosiran was well tolerated across ascending doses. Injection-site reactions were the only adverse events occurring in ≥10% of rapirosiran-treated participants (11%), were mild, and resolved transiently without intervention. No treatment-related serious adverse events were reported. Plasma concentrations of rapirosiran declined rapidly within 24 hours post-dose, and urinary excretion ranged between 17% and 37% across doses.

In Part B (MASH patients), the most common adverse event occurring in ≥10% of rapirosiran recipients was COVID-19 infection (14%; 5/36 cases), all deemed unrelated to the study drug. Importantly, no cases of drug-induced liver injury were observed in either study part, affirming the hepatic safety profile of rapirosiran.

Fig. 3

Fig. 3 HSD17B13 mRNA percentage change in liver biopsies at Month 6 and Month 12 in patients with metabolic dysfunction-associated steatohepatitis.
Fig. 4

Fig. 4 ALT levels over time in patients with metabolic dysfunction-associated steatohepatitis.
Fig. 5

Fig. 5 AST levels over time in patients with metabolic dysfunction-associated steatohepatitis.

Pharmacodynamics
Rapirosiran demonstrated a potent, dose-dependent inhibition of HSD17B13 mRNA expression in liver biopsy specimens obtained post-treatment. The highest-dose group (400 mg) showed a median reduction of 78% in liver HSD17B13 mRNA at 6 months compared to baseline, signaling effective target engagement in the MASH patient population.

Fig. 6

Fig. 6 Liver histology changes in patients with metabolic dysfunction-associated steatohepatitis.

The magnitude and durability of HSD17B13 suppression align with the mechanism of RNAi therapeutics and have important implications for ameliorating pathogenic processes in MASH.

Expert Commentary

This study validates the translational approach of leveraging human genetics to guide RNAi drug development in chronic liver disease. The favorable safety profile of rapirosiran in both healthy volunteers and patients with MASH is especially promising because many investigational agents in this field have encountered adverse hepatic or systemic effects.

Moreover, the achievement of substantial hepatic target mRNA knockdown with only two doses spaced 12 weeks apart reflects the pharmacological advantages of GalNAc-conjugated siRNAs, which facilitate hepatocyte-specific delivery and prolonged activity.

However, this phase I study is limited by its relatively small sample size and short duration. While robust on-target pharmacodynamic effects were observed, clinical efficacy in terms of histological improvement, fibrosis regression, or biochemical markers was not captured and remains to be demonstrated in future trials.

The absence of drug-induced liver injury is reassuring but longer-term safety monitoring will be essential given the chronic nature of MASH treatment.

Conclusion

Rapirosiran represents an innovative RNAi therapeutic targeting HSD17B13, a gene genetically linked to reduced risk of chronic liver disease. This phase I randomized, double-blind, placebo-controlled trial demonstrates that rapirosiran is safe, well tolerated, and effectively reduces hepatic HSD17B13 mRNA expression in adults with MASH.

These promising results lay the groundwork for subsequent phase II/III studies assessing rapirosiran’s clinical efficacy and long-term safety. Given the limited treatment landscape for MASH, rapirosiran offers a novel mechanism to modify disease pathogenesis and potentially improve patient outcomes.

Ongoing and future investigations will determine whether the potent gene silencing achieved translates into meaningful therapeutic benefits in MASH and related metabolic liver diseases.

References

1. Sanyal AJ, Taubel J, Badri P, et al. Phase I randomized double-blind study of an RNA interference therapeutic targeting HSD17B13 for metabolic dysfunction-associated steatohepatitis. J Hepatol. 2025 Oct;83(4):838-848. doi: 10.1016/j.jhep.2025.05.031 IF: 33.0 Q1 . Epub 2025 Jun 27. PMID: 40581300 IF: 33.0 Q1 .

2. Abul-Husn NS, Cheng X, Li AH, et al. A Protein-Truncating HSD17B13 Variant and Protection from Chronic Liver Disease. N Engl J Med. 2018;378(12):1096-1106. doi:10.1056/NEJMoa1702192 .

3. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global Epidemiology of Nonalcoholic Fatty Liver Disease—Meta-Analytic Assessment of Prevalence, Incidence, and Outcomes. Hepatology. 2016;64(1):73-84. doi:10.1002/hep.28431 IF: 15.8 Q1 .

Comments

No comments yet. Why don’t you start the discussion?

Leave a Reply

Your email address will not be published. Required fields are marked *