MedPath

DCR-CM2 Advanced Drug Monograph

Published:Jun 4, 2025

Generic Name

DCR-CM2

Comprehensive Report on DCR-CM2 (Lepodisiran; LY3819469): An Investigational siRNA Therapeutic for Elevated Lipoprotein(a)

I. Introduction to DCR-CM2 (Lepodisiran)

A. Overview of DCR-CM2 (Lepodisiran) as an Investigational Therapeutic

Lepodisiran, identified by the development codes DCR-CM2 and LY3819469, is an investigational therapeutic agent belonging to the class of small interfering RNAs (siRNAs).[1] It is specifically engineered for the treatment of hyperlipidemia, with a primary focus on reducing elevated plasma concentrations of lipoprotein(a) [Lp(a)].[1] Lp(a) is a genetically determined lipoprotein particle recognized as an independent causal risk factor for atherosclerotic cardiovascular disease (ASCVD), calcific aortic valve stenosis, and overall cardiovascular morbidity and mortality.[1] Elevated levels of Lp(a) are prevalent, affecting an estimated 20-25% of the global population.[4] The existence of multiple development codes, DCR-CM2 and LY3819469, likely reflects the progression of the drug's development, originating from Dicerna Pharmaceuticals (where "DCR-" prefixes are common for pipeline candidates) and subsequently advancing under Eli Lilly and Company (where "LY" prefixes are characteristic).[13] This transition from a smaller biotechnology company, often specializing in innovative platform technologies, to a large pharmaceutical firm for extensive late-stage clinical trials and potential commercialization is a well-established pattern in the pharmaceutical industry, particularly for therapeutics targeting large patient populations and requiring substantial investment in outcomes studies.

B. Synonyms, Chemical Class, and Key Identifiers

Lepodisiran is also known by its alternative development codes, LY3819469 and DCR-CM2.[1] Chemically, it is classified as a small interfering RNA (siRNA).[1] More specifically, Lepodisiran is an N-acetylgalactosamine (GalNAc)-conjugated siRNA, a modification designed to facilitate targeted delivery to hepatocytes (liver cells).[2] Key chemical and regulatory identifiers for Lepodisiran are summarized in Table 1.

Table 1: DCR-CM2 (Lepodisiran) - Overview

CharacteristicDetailReferences
Drug NameLepodisiran1
Development CodesDCR-CM2, LY38194691
Chemical ClassGalNAc-conjugated Small Interfering RNA (siRNA)2
DevelopersEli Lilly and Company (Current); Dicerna Pharmaceuticals (Originator)2
CAS Number2808361-18-23
UNIIX4BE7S8PN93
WHO Number1306819
Therapeutic TargetLipoprotein(a) [Lp(a)] via apolipoprotein(a) [apo(a)] mRNA1

This consolidated overview provides essential identifiers and foundational information for Lepodisiran, ensuring clarity and context for the subsequent detailed discussion.

C. Developers and Collaborators

Lepodisiran is currently undergoing late-stage clinical development under the stewardship of Eli Lilly and Company.[2] The drug candidate was originally discovered and developed by Dicerna Pharmaceuticals, leveraging their proprietary GalXC™ RNAi technology platform, through a collaborative agreement with Eli Lilly.[1] Subsequently, Eli Lilly licensed the drug and assumed lead responsibility for its global development and potential commercialization.[20] The assumption of lead development by a major pharmaceutical entity like Eli Lilly signifies a substantial commitment and underscores the perceived therapeutic potential of Lepodisiran. The development of RNAi-based therapies, especially for prevalent conditions such as ASCVD, necessitates extensive and costly Phase 3 cardiovascular outcome trials, a scale of endeavor typically undertaken by large pharmaceutical companies with established global infrastructure and resources for such large-scale programs. Dicerna's strategy has often involved partnering its GalXC™ platform assets for development in non-core areas or where extensive late-stage development is required.[13]

D. Target Indication: Elevated Lipoprotein(a) [Lp(a)] and Atherosclerotic Cardiovascular Disease (ASCVD)

The primary therapeutic indication for Lepodisiran is the reduction of elevated Lp(a) levels in individuals at risk for ASCVD.[1] Lp(a) is a unique lipoprotein particle whose plasma concentration is predominantly determined by genetic factors, specifically variations in the LPA gene encoding apolipoprotein(a).[9] Elevated Lp(a) concentrations are recognized as an independent, causal risk factor for a spectrum of cardiovascular pathologies, including myocardial infarction, ischemic stroke, peripheral artery disease, and calcific aortic valve stenosis.[1] A critical aspect of Lp(a) as a risk factor is that its levels are not significantly modulated by lifestyle interventions or by most currently available lipid-lowering therapies, such as statins or ezetimibe; PCSK9 inhibitors have shown only modest Lp(a) reduction.[4] This creates a significant unmet medical need for therapies that can effectively and specifically lower Lp(a) concentrations. The development of Lepodisiran represents a novel therapeutic strategy that moves beyond conventional cholesterol management, primarily focused on LDL-cholesterol (LDL-C), to address the residual cardiovascular risk conferred by this distinct genetic factor. By targeting Lp(a), Lepodisiran aims to fill a critical gap in cardiovascular prevention, particularly for individuals who remain at high risk despite optimal management of other modifiable risk factors.

II. Mechanism of Action and Pharmacological Profile

A. Detailed Explanation of siRNA-mediated Interference (RNAi)

Lepodisiran exerts its pharmacological effect through the mechanism of RNA interference (RNAi), a natural cellular process that regulates gene expression.[1] siRNAs are short, double-stranded RNA (dsRNA) molecules, typically 20-25 base pairs in length, that can induce sequence-specific silencing of gene expression at the post-transcriptional level.[1] Upon introduction into the cytoplasm, the dsRNA is processed, and its antisense strand (or guide strand) is incorporated into a multi-protein complex known as the RNA-Induced Silencing Complex (RISC).[17] The RISC, now programmed by the antisense strand, seeks out and binds to messenger RNA (mRNA) molecules that possess a complementary sequence.[17] This binding event, mediated by Watson-Crick base pairing, leads to the endonucleolytic cleavage of the target mRNA by Argonaute-2, a key enzymatic component of RISC.[17] The cleaved mRNA is subsequently degraded by cellular nucleases, thereby preventing its translation into the corresponding protein.[1] The inherent specificity of this process, dictated by the nucleotide sequence of the siRNA, allows for highly targeted gene silencing. This precision offers a potential advantage over traditional small molecule drugs, which may have broader target profiles and a higher likelihood of off-target interactions, by minimizing unintended pharmacological effects, provided the siRNA is carefully designed to avoid significant homology with non-target mRNAs.[18]

B. Specific Targeting of Apolipoprotein(a) [apo(a)] mRNA

Lepodisiran is engineered with a specific nucleotide sequence designed to target the mRNA that encodes apolipoprotein(a) [apo(a)].[1] Apo(a) is a large, polymorphic glycoprotein that is covalently linked to apolipoprotein B-100 (apoB-100) within an LDL-like particle, forming the complete Lp(a) particle.[1] The synthesis of apo(a) in the liver is the rate-limiting step in the biogenesis of Lp(a) particles.[6] By specifically targeting apo(a) mRNA for degradation, Lepodisiran aims to inhibit the production of the apo(a) protein, thereby directly interfering with the formation of new Lp(a) particles. This upstream mechanism of action, preventing the synthesis of a key structural component, is a more direct approach to lowering Lp(a) levels than strategies that might attempt to enhance the clearance of already formed particles or modulate their atherogenic properties, which could be less efficient or face other biological limitations.

C. Consequent Reduction of Lp(a) Synthesis and Plasma Levels

Following the RISC-mediated degradation of apo(a) mRNA within hepatocytes (the primary site of Lp(a) synthesis), the intracellular pool of apo(a) protein available for Lp(a) particle assembly is diminished.[1] This reduction in apo(a) protein synthesis directly leads to a decreased rate of formation and secretion of Lp(a) particles from the liver into the bloodstream. Consequently, the plasma concentration of Lp(a) is lowered.[1] Extensive clinical trial data from Phase 1 and Phase 2 studies have consistently demonstrated that administration of Lepodisiran results in substantial, dose-dependent, and durable reductions in plasma Lp(a) levels.[4] The magnitude of Lp(a) reduction, often exceeding 90% with higher doses, underscores the high efficiency of the RNAi mechanism and the effectiveness of the GalNAc-mediated delivery system in reaching the target hepatocytes and engaging the cellular machinery for gene silencing. Such profound lowering of a target protein is rarely achieved with conventional oral lipid-modifying therapies and highlights the potency of this therapeutic approach.

III. The GalXC™ RNAi Technology Platform

A. Overview of Dicerna's GalXC™ and GalXC-Plus™ Technology

Lepodisiran's development is rooted in Dicerna Pharmaceuticals' proprietary GalXC™ RNAi technology platform.[2] The GalXC™ platform is a patented and advanced RNAi technology specifically engineered to generate siRNA molecules with optimized pharmaceutical properties, including enhanced potency, stability, and targeted delivery.[13] A key feature of this platform is the conjugation of N-acetylgalactosamine (GalNAc) ligands to the siRNA duplex.[13] The GalXC-Plus™ technology represents a further evolution, aiming to extend RNAi delivery capabilities beyond the liver to other tissues, such as the central nervous system (CNS).[23] However, for Lepodisiran, which targets the liver-synthesized apo(a), the liver-targeting attributes of the core GalXC™ technology are paramount. The platform's strength lies in its ability to produce siRNA therapeutics that are suitable for subcutaneous administration, exhibit a long duration of action, demonstrate high target specificity, and possess a potentially favorable therapeutic index, all of which are critical for a drug like Lepodisiran.[13]

B. Mechanisms for Liver Targeting (GalNAc Conjugation, ASGPR Interaction)

The GalXC™ platform achieves liver-specific targeting through the strategic conjugation of GalNAc ligands, typically in a trivalent configuration, to the sense strand of the siRNA molecule.[13] These GalNAc sugar moieties have a high and specific binding affinity for the asialoglycoprotein receptor (ASGPR), a C-type lectin that is abundantly and almost exclusively expressed on the sinusoidal surface of hepatocytes.[17] Upon subcutaneous administration and entry into systemic circulation, the GalNAc-siRNA conjugate encounters ASGPR on hepatocytes. The interaction between GalNAc and ASGPR triggers receptor-mediated endocytosis, leading to the efficient internalization of the siRNA specifically into liver cells.[17] This highly specific uptake mechanism is a cornerstone of the technology, as it concentrates the therapeutic payload in the target organ (liver) while minimizing systemic exposure and potential off-target effects in other tissues. Such selective biodistribution is anticipated to enhance the drug's safety profile and therapeutic index.

C. Features Enabling Subcutaneous Delivery and Long Duration of Action

GalXC™ siRNAs are meticulously engineered for enhanced stability and an extended pharmacodynamic effect. This is achieved through various chemical modifications incorporated into the siRNA structure, such as phosphorothioate linkages in the backbone and 2'-O-methyl or other 2'-sugar modifications.[16] These modifications serve to protect the siRNA molecule from degradation by endo- and exonucleases present in the bloodstream and within cells, thereby prolonging its biological half-life and contributing to a sustained duration of action.[16] The GalXC™ platform is designed to yield therapeutics suitable for subcutaneous administration, which offers greater convenience for patients compared to intravenous infusions, particularly for chronic therapies.[2] The combination of efficient liver uptake via the GalNAc-ASGPR pathway and the enhanced intracellular stability of the siRNA, particularly once loaded into the RISC complex within long-lived hepatocytes, results in a remarkably prolonged pharmacodynamic effect. This allows for infrequent dosing schedules, with administrations potentially spaced several months apart (e.g., every 3-6 months or even less frequently), as suggested by the sustained Lp(a) reduction observed with Lepodisiran.[4] The impressive duration of action seen with Lepodisiran, where significant Lp(a) suppression is maintained for many months (Phase 1 data showed effects for up to 48 weeks after a single dose [25], and Phase 2 data indicated sustained reductions for nearly 1.5 years with two doses [4]), is a direct consequence of these engineered platform attributes. This characteristic is a major clinical advantage for the management of chronic conditions, promoting better patient adherence.

IV. Preclinical Development Highlights

While specific, detailed preclinical study reports for Lepodisiran (DCR-CM2/LY3819469) itself are not extensively covered in the provided information, the capabilities of the underlying GalXC™ platform and data from closely related molecules provide strong supportive evidence. Dicerna's GalXC platform has consistently demonstrated potent gene silencing, often exceeding 90% target mRNA knockdown in non-human primates (NHPs) after a single subcutaneous dose across various gene targets.34 For instance, a single 3 mg/kg dose of a GalXC compound achieved 94% silencing of one undisclosed rare disease gene target and 97% silencing of another in NHPs, highlighting the platform's efficacy.34

Furthermore, preclinical studies with SLN360, another GalNAc-conjugated siRNA that also targets LPA mRNA (the same gene target as Lepodisiran), demonstrated specific LPA mRNA reduction of up to 91% and potent serum Lp(a) protein reduction of up to 95% in cynomolgus monkeys. These effects were long-lasting, persisting for at least 9 weeks.18 The safety profile in these NHP studies was consistent with known effects of the GalNAc-siRNA platform, including reversible, non-adverse microscopic changes in the kidney (related to siRNA clearance) and liver (related to exaggerated pharmacology or high intracellular siRNA concentrations).18 These findings are generally considered non-adverse at therapeutic exposure levels and are well-characterized for this class of compounds.

The robust preclinical efficacy, characterized by a high degree of target knockdown and a prolonged duration of action, coupled with efficient liver targeting demonstrated by the GalXC platform across multiple gene targets, including LPA mRNA, provided a compelling scientific rationale for advancing Lepodisiran into human clinical trials. The predictable, platform-related safety signals observed in preclinical toxicology studies also helped to inform the design of early clinical trials, including dose selection and safety monitoring plans.

V. Clinical Development Program

A. Phase 1 Studies (e.g., NCT04914546)

The initial human evaluation of Lepodisiran was conducted in a Phase 1 clinical trial (NCT04914546) designed to assess its safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD).[25]

  • Study Design, Objectives, and Participant Characteristics: This was a randomized, double-blind, placebo-controlled, single ascending-dose study involving 48 adult participants.25 Eligible individuals had baseline Lp(a) concentrations of at least 75 nmol/L (or ≥30 mg/dL) and did not have clinically manifest cardiovascular disease at enrollment.25 Participants were randomized to receive a single subcutaneous injection of Lepodisiran at one of six dose levels (4 mg, 12 mg, 32 mg, 96 mg, 304 mg, or 608 mg) or placebo.25 The primary objectives were to evaluate the safety and tolerability of Lepodisiran. Secondary objectives included characterization of its plasma PK profile and assessment of its effect on fasting Lp(a) concentrations over a follow-up period of up to 48 weeks.25
  • Key Safety, Tolerability, Pharmacokinetic (PK), and Pharmacodynamic (PD) Findings:
  • Safety and Tolerability: Lepodisiran was generally well-tolerated across all dose levels. The incidence of adverse events (AEs) was low, and no clear dose-related trend in AEs was observed.[25] Commonly reported AEs included headache, COVID-19 infection (background incidence), rhinorrhea, and ECG patch erythema.[25] Injection site reactions were also reported, with one source noting they were less frequent in the active treatment arms compared to the placebo arm [25], while another abstract of the same study mentioned one serious adverse event, though further details were not provided.[32]
  • Pharmacokinetics: Following subcutaneous administration, plasma concentrations of Lepodisiran reached peak levels (Tmax) within approximately 10.5 hours. The drug was rapidly cleared from the plasma, with levels becoming undetectable by 48 hours post-dose.[32] This PK profile, featuring rapid absorption and swift plasma clearance, is characteristic of GalNAc-siRNA therapeutics and is indicative of efficient uptake into the target liver tissue, where the drug exerts its prolonged effect.
  • Pharmacodynamics (Lp(a) Reduction): Lepodisiran demonstrated a potent, dose-dependent reduction in serum Lp(a) concentrations. The effect was notably durable, with the highest single dose tested (608 mg) resulting in a median Lp(a) reduction of 94% at 48 weeks (Day 337) post-administration.[25] Maximal median Lp(a) reductions varied by dose, ranging from -41% in the 4 mg group to -97% in the 608 mg group at earlier time points.[32] The profound and sustained Lp(a) lowering observed after a single dose provided strong proof-of-concept for Lepodisiran's therapeutic potential.

Table 2: Summary of Lepodisiran Phase 1 Clinical Trial (NCT04914546) Design and Key Findings

CharacteristicDetailReferences
Trial IDNCT0491454625
Phase125
DesignRandomized, Double-Blind, Placebo-Controlled, Single Ascending Dose25
No. of Participants4825
Key Inclusion CriteriaAdults, Lp(a) ≥75 nmol/L (or ≥30 mg/dL), no known CVD25
Dose RangesSingle SC doses: 4 mg, 12 mg, 32 mg, 96 mg, 304 mg, 608 mg, or placebo25
Primary OutcomeSafety and Tolerability25
Key PD OutcomeMedian Lp(a) reduction of 94% at 48 weeks with 608 mg dose25
Key PK FindingTmax ~10.5 hours; plasma levels undetectable by 48 hours32

The dissociation between the short plasma pharmacokinetic half-life (undetectable by 48 hours) and the very long pharmacodynamic duration of action (Lp(a) reduction sustained for at least 48 weeks) is a defining feature of GalNAc-siRNA therapeutics. This phenomenon is attributed to the rapid and efficient uptake of the siRNA into hepatocytes, followed by its stable incorporation into the RISC complex, enabling prolonged intracellular activity long after the drug has been cleared from systemic circulation.[17]

B. Phase 2 Study – The ALPACA Trial (NCT05565742)

Following the promising Phase 1 results, Lepodisiran advanced to a Phase 2b trial, known as the ALPACA study (NCT05565742), to further evaluate its efficacy, safety, and dosing regimens in a larger population with elevated Lp(a).[4]

  • Detailed Study Design: The ALPACA trial was a multicenter, international, randomized, double-blind, placebo-controlled study.4 A total of 320 participants were enrolled and randomized in a 1:2:2:2:2 ratio into five treatment arms, receiving subcutaneous injections at baseline and again at Day 180 4:
  • Arm 1: Placebo at baseline and Day 180 (n=69) [4]
  • Arm 2: Lepodisiran 16 mg at baseline and Day 180 (n=36) [4]
  • Arm 3: Lepodisiran 96 mg at baseline and Day 180 (n=74) [4]
  • Arm 4: Lepodisiran 400 mg at baseline and Day 180
  • Arm 5: Lepodisiran 400 mg at baseline and Placebo at Day 180 For the primary analysis, data from the two arms receiving 400 mg of Lepodisiran at baseline (Arms 4 and 5) were pooled (total n=141 for pooled 400 mg group).[4] The study duration extended up to 540 days (approximately 1.5 years) to assess the durability of effect.[4] The trial was conducted at 66 sites across North America, Europe, Asia, and other regions.[8]
  • Participant Demographics/Baseline Characteristics: Participants were adults aged 40 years or older with a baseline serum Lp(a) concentration of at least 175 nmol/L.6 The median baseline Lp(a) level across the study population was 253.9 nmol/L.10 A significant proportion of participants were on concomitant stable doses of lipid-modifying therapies for at least 4 weeks prior to screening, including statins (74%), ezetimibe (33%), and PCSK9 inhibitors (6%).27 Reflecting a population at cardiovascular risk, 68% were deemed at high risk for a cardiovascular event, 48% had established coronary artery disease, and 31% had a history of previous myocardial infarction.30
  • Inclusion/Exclusion Criteria: Key inclusion criteria included age ≥40 years and serum Lp(a) concentration ≥175 nmol/L.27 Participants on stable lipid-modifying drugs were permitted. Major exclusion criteria comprised a cardiovascular event within 3 months prior to screening, moderate or severe heart failure, significant renal impairment (estimated Glomerular Filtration Rate <30 mL/min/1.73 m2), or evidence of significant liver disease (hepatic enzyme levels >3 times the upper limit of normal [ULN]).27 Women of child-bearing potential were also excluded.27
  • Primary and Key Secondary Endpoints: The primary endpoint was the placebo-adjusted, time-averaged percent change in serum Lp(a) concentration from baseline during the period from Day 60 to Day 180.4 Key secondary endpoints included the assessment of Lp(a) reduction at various other time intervals and specific time points up to Day 540, changes in other lipid parameters such as apolipoprotein B (ApoB), LDL-cholesterol (LDL-C), and inflammatory markers like high-sensitivity C-reactive protein (hs-CRP), as well as the percentage of participants achieving Lp(a) concentrations below specific thresholds.4
  • Comprehensive Efficacy Results: Lepodisiran demonstrated robust and dose-dependent efficacy in lowering Lp(a) levels 4:
  • Lp(a) Reduction (Primary Endpoint: Placebo-adjusted time-averaged % change, Day 60-180):
  • 16 mg Lepodisiran: -40.8 percentage points (95% Confidence Interval [CI], -55.8 to -20.6)
  • 96 mg Lepodisiran: -75.2 percentage points (95% CI, -80.4 to -68.5)
  • 400 mg Lepodisiran (pooled baseline doses): -93.9 percentage points (95% CI, -95.1 to -92.5)
  • Durability of Lp(a) Reduction (400 mg Lepodisiran at baseline and Day 180 arm):
  • The placebo-adjusted time-averaged percent change from baseline in Lp(a) from Day 30 to Day 360 was -94.8 percentage points.
  • At Day 360 (approximately 1 year), Lp(a) levels remained 91.0% below baseline.
  • At Day 540 (approximately 1.5 years), Lp(a) levels remained 74.2% below baseline.
  • Durability of Lp(a) Reduction (400 mg Lepodisiran at baseline, Placebo at Day 180 arm):
  • The placebo-adjusted time-averaged percent change from baseline in Lp(a) from Day 30 to Day 360 was -88.5 percentage points.
  • Apolipoprotein B (ApoB) Reduction (400 mg Lepodisiran dose):
  • Reductions from baseline of 14.1% at Day 60 and 13.7% at Day 180 were observed.
  • In the arm receiving a second 400 mg dose at Day 180, ApoB reductions were sustained through Day 540. The impressive durability of Lp(a) reduction, with substantial effects lasting up to 1.5 years after only two 400 mg doses, strongly supports the potential for an infrequent maintenance dosing schedule, such as biannually. This would be a considerable advantage for patient adherence in a long-term preventive care setting. Furthermore, the observed reduction in ApoB levels, while more modest than the Lp(a) lowering, is a positive secondary finding. As ApoB reflects the total concentration of atherogenic lipoproteins, this effect could contribute to a broader improvement in the atherogenic lipid profile, potentially enhancing overall cardiovascular risk reduction.
  • Full Safety and Tolerability Data: The safety profile of Lepodisiran in the ALPACA trial was generally favorable 4:
  • Treatment-Emergent Adverse Events (TEAEs) related to study drug:
  • Placebo group: 1% (1 of 69 participants)
  • 16 mg Lepodisiran group: 3% (1 of 36 participants)
  • 96 mg Lepodisiran group: 12% (9 of 74 participants)
  • Pooled 400 mg Lepodisiran group: 14% (20 of 141 participants)
  • Injection Site Reactions (ISRs): These were the most common drug-related AEs, occurring in a dose-dependent manner. They were generally mild in severity. In the highest dose (400 mg) group, ISRs were reported in up to 11.6-12% of participants, compared to 1% in the placebo group.[24]
  • Serious Adverse Events (SAEs): A total of 35 participants (11%) experienced SAEs during the study; however, none of these were deemed by investigators to be related to Lepodisiran or placebo treatment.[4]
  • Deaths: There was one death reported in the 16 mg Lepodisiran group, attributed to complications of chronic coronary disease/cardiomyopathy; this event was not considered related to the study drug.[4]
  • Withdrawals due to TEAEs: One participant in the placebo group withdrew from the study due to a TEAE. Importantly, no participants receiving Lepodisiran withdrew from treatment or the study due to TEAEs.[4]
  • Liver Function Tests (LFTs): Clinically meaningful elevations of alanine aminotransferase (ALT) or aspartate aminotransferase (AST) (defined as >3 times ULN) were observed in 2.7% to 5.8% of participants across the Lepodisiran intervention arms.[27] These elevations were generally transient. The safety findings from ALPACA are encouraging for a novel therapeutic. The absence of drug-related SAEs and no discontinuations due to AEs in the Lepodisiran arms are positive indicators of its tolerability. ISRs and transient LFT elevations are known class effects for subcutaneously administered GalNAc-siRNA therapeutics and will require continued monitoring in Phase 3 studies, but the observed frequencies appear within an acceptable range for this class of drugs.

Table 3: Lepodisiran Phase 2 ALPACA Trial (NCT05565742) – Study Design and Endpoints

CharacteristicDetailReferences
Trial IDNCT055657424
Phase2b4
DesignRandomized, Double-Blind, Placebo-Controlled, Multi-dose, Multicenter, International4
No. of Participants3204
Arms/Dosing RegimensPlacebo SC; Lepodisiran 16mg SC; Lepodisiran 96mg SC; Lepodisiran 400mg SC (all at baseline & Day 180); Lepodisiran 400mg SC then Placebo4
DurationUp to 540 days4
Key Inclusion CriteriaAdults ≥40 years, Serum Lp(a) ≥175 nmol/L6
Primary EndpointPlacebo-adjusted time-averaged % change in Lp(a) from baseline (Day 60-180)4
Key Secondary EndpointsLp(a) durability (up to Day 540), ApoB changes, LDL-C changes, hs-CRP changes, Safety and Tolerability4

C. Phase 3 Study – The ACCLAIM-Lp(a) Trial (NCT06292013)

Building on the positive Phase 2 results, Lepodisiran has progressed to a large-scale Phase 3 cardiovascular outcomes trial named ACCLAIM-Lp(a) (NCT06292013).[2]

  • Study Design and Objectives: The ACCLAIM-Lp(a) trial is a Phase 3, randomized, double-blind, placebo-controlled study designed to definitively evaluate the efficacy and safety of Lepodisiran in reducing the risk of Major Adverse Cardiovascular Events (MACE) in adults with elevated Lp(a) levels who have established ASCVD or are at high risk for a first cardiovascular event.4
  • Primary Endpoint: The primary endpoint of the ACCLAIM-Lp(a) trial is the time to first occurrence of a composite of MACE.41 While the specific components of the MACE composite for this trial are not fully detailed in all available sources, MACE in cardiovascular outcome trials typically includes cardiovascular death, nonfatal myocardial infarction, and nonfatal stroke.45 Some trials may also include components like coronary revascularization or unstable angina requiring hospitalization.46 The trial will assess whether treatment with Lepodisiran leads to a statistically significant reduction in these events compared to placebo.4 The critical objective of this trial is to determine if the profound Lp(a) lowering observed with Lepodisiran translates into a tangible clinical benefit in terms of preventing cardiovascular events, thereby validating the "Lp(a) hypothesis."
  • Estimated Enrollment, Key Inclusion/Exclusion Criteria:
  • Estimated Enrollment: The trial aims to enroll approximately 12,500 participants globally.[41]
  • Key Inclusion Criteria (summarized from [41]):
  • Participants must be adults (age ≥18 years).
  • Elevated Lp(a) levels, specified as ≥175 nmol/L (another source mentions ≥200 nmol/L [9]).
  • Participants must meet criteria for either:
  • Established ASCVD (e.g., history of myocardial infarction, prior coronary revascularization with at least one additional cardiovascular risk factor).
  • High-risk primary prevention: Individuals aged ≥55 years who are at risk for a first cardiovascular event and have either documented coronary artery disease (CAD), carotid stenosis, or peripheral artery disease (PAD) without a history of a major event or revascularization; or known familial hypercholesterolemia; or a combination of other high-risk factors.
  • Key Exclusion Criteria (summarized from [41]):
  • Recent (within 90 days of screening) or planned lipoprotein apheresis during the study.
  • Severe renal failure (eGFR <15 mL/min/1.73$m^2$) or current dialysis.
  • Diagnosis of active nephrotic syndrome or very high urine albumin-to-creatinine ratio (UACR ≥5000 mg/g).
  • Evidence of acute or chronic hepatitis, known cirrhosis, signs and symptoms of any other liver disease (excluding metabolic-associated steatotic liver disease), or exclusionary liver function laboratory results at screening. The inclusion of both secondary prevention (established ASCVD) and high-risk primary prevention cohorts in the ACCLAIM-Lp(a) trial suggests an ambition to demonstrate broad utility for Lepodisiran. Positive outcomes across these diverse patient groups could support a comprehensive therapeutic indication, potentially transforming the management of Lp(a)-mediated cardiovascular risk for a wide spectrum of at-risk individuals.
  • Study Timelines and Locations:
  • Study Start Date: March 2024.[41]
  • Estimated Primary Completion Date: March 2029.[2]
  • Locations: The trial is a multicenter, international effort with sites in various countries, including the USA (e.g., Stanford Healthcare, University of California San Diego) and Europe (e.g., Czechia).[41]

Table 4: Lepodisiran Phase 3 ACCLAIM-Lp(a) Trial (NCT06292013) – Overview and Primary Outcome

CharacteristicDetailReferences
Trial IDNCT0629201341
Phase32
DesignRandomized, Double-Blind, Placebo-Controlled, Cardiovascular Outcomes Trial41
ObjectiveTo evaluate the effect of Lepodisiran on the reduction of Major Adverse Cardiovascular Events (MACE)4
Estimated EnrollmentApprox. 12,500 participants41
Key PopulationAdults with elevated Lp(a) (≥175 nmol/L) and established ASCVD or at high risk for a first cardiovascular event41
Primary EndpointTime to first occurrence of a composite of MACE (typically cardiovascular death, nonfatal MI, nonfatal stroke; specific components TBD)[41 (general MACE)]
Estimated Completion DateMarch 20292

D. Studies in Special Populations (e.g., Hepatic Impairment - NCT06916078 / NCT0591635)

To understand the behavior of Lepodisiran in specific patient subgroups, a dedicated Phase 1 study (identified as NCT06916078 and also referred to as NCT0591635 15) is being conducted. This trial aims to evaluate the pharmacokinetics, safety, and tolerability of Lepodisiran in participants with varying degrees of liver function—normal, mild, moderate, or severe hepatic impairment—compared to individuals with normal hepatic function.15 Participants in this study receive a single subcutaneous injection of Lepodisiran. The study involves a relatively short duration of participation (approximately 9 weeks), which includes an in-patient residence period (5 days/4 nights) for intensive monitoring and subsequent outpatient follow-up visits.47 Key pharmacokinetic parameters being assessed include the area under the plasma concentration-time curve (AUC) and the maximum plasma concentration (Cmax) of Lepodisiran.48

Given that Lepodisiran is a GalNAc-conjugated siRNA specifically targeted to the liver and that Lp(a) is synthesized hepatically, understanding its disposition and safety profile in patients with pre-existing liver conditions is of paramount importance. Hepatic impairment can alter drug metabolism, distribution, and the expression levels of receptors like ASGPR, all of which could potentially influence the efficacy and safety of Lepodisiran. The data from this study will be critical for developing appropriate dosing recommendations and for identifying any specific risks or precautions needed when administering Lepodisiran to patients with compromised liver function. This is a standard and necessary component of comprehensive drug development for any therapeutic that is primarily cleared by or acts upon the liver.

VI. Comprehensive Efficacy Analysis

A. In-depth Review of Lp(a) Lowering Across Studies and Dose Ranges

The efficacy of Lepodisiran in reducing plasma Lp(a) concentrations has been consistently demonstrated across its Phase 1 and Phase 2 clinical trials, exhibiting a clear dose-response relationship.

  • Phase 1 (NCT04914546): In this single ascending-dose study, Lepodisiran produced dose-dependent reductions in Lp(a). A single subcutaneous administration of the highest dose tested, 608 mg, resulted in a profound median Lp(a) reduction of 94% at 48 weeks (Day 337) post-dose.[25] Maximal median changes in Lp(a) concentration from baseline varied with dose, from -41% in the 4 mg group up to -97% in the 608 mg group at earlier assessment time points.[32]
  • Phase 2 (ALPACA Trial, NCT05565742): The multi-dose ALPACA trial further confirmed and characterized the potent Lp(a)-lowering effects of Lepodisiran.[4]
  • The primary endpoint, defined as the placebo-adjusted time-averaged percent change in Lp(a) from baseline over the period of Day 60 to Day 180, showed the following results:
  • 16 mg Lepodisiran dose group: -40.8% reduction.
  • 96 mg Lepodisiran dose group: -75.2% reduction.
  • 400 mg Lepodisiran dose group (pooled analysis of participants receiving 400 mg at baseline): -93.9% reduction. These findings from both early and mid-stage clinical development consistently highlight Lepodisiran's capacity to achieve substantial reductions in Lp(a) levels.

B. Effects on Other Relevant Biomarkers

Beyond its primary effect on Lp(a), the ALPACA trial also assessed Lepodisiran's impact on other relevant cardiovascular biomarkers:

  • Apolipoprotein B (ApoB): In the ALPACA trial, participants receiving the 400 mg dose of Lepodisiran exhibited reductions in ApoB levels from baseline by 14.1% at Day 60 and 13.7% at Day 180. For participants who received a second 400 mg dose at Day 180, these ApoB reductions were sustained through Day 540.[4]
  • LDL-Cholesterol (LDL-C) and High-Sensitivity C-Reactive Protein (hs-CRP): Changes in LDL-C and hs-CRP were included as secondary endpoints in the ALPACA trial design.[31] However, specific quantitative results for the impact of Lepodisiran on LDL-C and hs-CRP are not consistently detailed in the available summarized materials. The consistent and profound Lp(a) reduction, coupled with a clear dose-response, strongly validates the drug's potent mechanism of action. The additional observation of ApoB reduction, although more modest in magnitude compared to Lp(a) lowering, is clinically relevant. ApoB is a structural protein found in all major atherogenic lipoproteins (LDL, VLDL, IDL, and Lp(a)); thus, a reduction in total ApoB may indicate a decrease in the overall burden of these atherogenic particles, potentially offering an incremental benefit to cardiovascular risk reduction beyond Lp(a) lowering alone.

C. Durability of Effect

A standout characteristic of Lepodisiran, evident from its clinical development program, is the exceptional durability of its Lp(a)-lowering effect:

  • Phase 1 (NCT04914546): A single 608 mg subcutaneous dose of Lepodisiran demonstrated sustained Lp(a) reduction for at least 337 days (48 weeks).[25]
  • Phase 2 (ALPACA Trial, NCT05565742):
  • In the arm receiving 400 mg of Lepodisiran at baseline and again at Day 180, Lp(a) levels remained profoundly suppressed, being 91.0% below baseline at Day 360 (approximately 1 year) and still 74.2% below baseline at Day 540 (approximately 1.5 years).[4]
  • Even with a single 400 mg baseline dose followed by placebo at Day 180, the time-averaged Lp(a) reduction from Day 30 to Day 360 was -88.5 percentage points.[10] This remarkable and sustained duration of action is a direct consequence of the GalXC™ platform's design, which enhances siRNA stability and ensures prolonged activity within hepatocytes. Such durability strongly supports the feasibility of an infrequent dosing schedule, potentially biannual for the 400 mg dose. For a chronic, lifelong condition like elevated Lp(a), this offers a significant practical advantage in terms of patient convenience and adherence compared to therapies requiring more frequent administration, which is crucial for long-term preventive efficacy.

Table 5: Key Efficacy Results from Phase 2 ALPACA Trial (Lp(a) and ApoB Reduction by Dose)

Dose Group (SC)Primary Endpoint: Lp(a) Reduction (Placebo-Adjusted Time-Averaged % Change, Day 60-180)Lp(a) Reduction at Day 360 (vs Baseline, 400mg-400mg arm)Lp(a) Reduction at Day 540 (vs Baseline, 400mg-400mg arm)ApoB Reduction at Day 180 (vs Baseline, 400mg dose)References
PlaceboBaseline comparatorN/AN/AN/A4
Lepodisiran 16 mg-40.8% (95% CI: -55.8 to -20.6)Not specified for this doseNot specified for this doseNot specified for this dose4
Lepodisiran 96 mg-75.2% (95% CI: -80.4 to -68.5)Not specified for this doseNot specified for this doseNot specified for this dose4
Lepodisiran 400 mg (pooled baseline)-93.9% (95% CI: -95.1 to -92.5)-91.0%-74.2%-13.7%4

VII. Detailed Safety and Tolerability Profile

A. Consolidated Overview of Adverse Events (Common, Serious) Across Clinical Trials

The safety and tolerability of Lepodisiran have been evaluated in Phase 1 and Phase 2 clinical trials.

  • Phase 1 (NCT04914546): Lepodisiran was reported to be generally well-tolerated. The overall rate of adverse events (AEs) was low, and there was no discernible dose-related trend in their occurrence.[25] The most frequently reported AEs included headache, COVID-19 infection (likely reflecting background community transmission), rhinorrhea, and ECG patch erythema.[25] Injection site reactions were also noted.[25] One abstract mentioned a single serious adverse event (SAE) in the study, but details regarding its nature and relationship to the study drug were not provided.[32]
  • Phase 2 (ALPACA Trial, NCT05565742): The safety profile in the larger, longer-duration ALPACA trial remained largely favorable.[4]
  • Crucially, no SAEs were deemed by investigators to be related to treatment with Lepodisiran.
  • One death occurred in a participant receiving the 16 mg dose of Lepodisiran; this was attributed to complications of chronic coronary disease/cardiomyopathy and was not considered related to the study drug.
  • No participants in any Lepodisiran treatment arm discontinued the study due to TEAEs. In contrast, one participant in the placebo arm withdrew due to a TEAE.

B. Injection Site Reactions (ISRs)

ISRs are a common occurrence with subcutaneously administered therapies, particularly oligonucleotides.

  • In the Phase 1 study, ISRs were reported.[25]
  • In the Phase 2 ALPACA trial, ISRs were observed to be dose-dependent and were generally mild in severity. They occurred in up to 11.6-12% of participants in the highest Lepodisiran dose group (400 mg), compared to a rate of 1% in the placebo group.[24]

C. Effects on Liver Function Tests (LFTs) and Other Notable Safety Signals

Given the liver-targeted nature of GalNAc-siRNA therapeutics, liver function is a key area of safety monitoring.

  • In the Phase 2 ALPACA trial, clinically meaningful elevations of liver enzymes (ALT or AST >3 times the ULN) were observed in a small percentage of participants in the Lepodisiran intervention arms, ranging from 2.7% to 5.8%.[27] These elevations were generally reported as transient.
  • This observation is consistent with findings from preclinical studies of similar GalNAc-siRNA compounds (e.g., SLN360), which showed reversible microscopic changes in the liver, considered non-adverse and platform-related effects at therapeutic exposures.[18]

D. Comparison Across Dose Groups and with Placebo (ALPACA Trial)

A direct comparison of TEAEs related to the study drug in the ALPACA trial reveals a dose-dependent increase, though overall rates remained relatively low:

  • Placebo group: 1% (1 of 69 participants)
  • 16 mg Lepodisiran group: 3% (1 of 36 participants)
  • 96 mg Lepodisiran group: 12% (9 of 74 participants)
  • Pooled 400 mg Lepodisiran group: 14% (20 of 141 participants).[4]

The occurrence of ISRs and transient LFT elevations are recognized as potential class effects for subcutaneously administered GalNAc-siRNA therapeutics. The frequencies observed for Lepodisiran in the ALPACA trial appear to be within an anticipated range for this class of drugs. While these events were generally mild and manageable, their characterization will be an important aspect of the larger and longer-duration Phase 3 ACCLAIM-Lp(a) trial. The overall tolerability profile from Phase 2, particularly the absence of drug-related SAEs or discontinuations due to AEs in the Lepodisiran arms, is a positive signal for a novel therapeutic intended for chronic use.

Table 6: Summary of Key Adverse Events in Phase 2 ALPACA Trial (Frequency by Dose Group vs. Placebo)

Adverse Event CategoryPlacebo (N=69)Lepodisiran 16 mg (N=36)Lepodisiran 96 mg (N=74)Lepodisiran 400 mg (Pooled, N=141)References
TEAEs Related to Study Drug (%)1% (1/69)3% (1/36)12% (9/74)14% (20/141)4
Any Serious Adverse Event (SAE) (%)(Overall 11% across study, none drug-related)(Overall 11% across study, none drug-related)(Overall 11% across study, none drug-related)(Overall 11% across study, none drug-related)4
Injection Site Reactions (%)1%8.1% - 11.6% (range across doses)8.1% - 11.6% (range across doses)up to 11.6-12%24
LFT Elevations (>3xULN, ALT or AST) (%)Not specified2.7% - 5.8% (range across intervention arms)2.7% - 5.8% (range across intervention arms)2.7% - 5.8% (range across intervention arms)27
Discontinuations due to Drug-Related AEs (%)0% (1 placebo withdrawal for TEAE)0%0%0%4

VIII. Pharmacokinetics and Pharmacodynamics of Lepodisiran

A. Absorption, Distribution, Metabolism, and Excretion (ADME) Profile

  • Absorption: Lepodisiran is administered via subcutaneous injection.[2] Following administration, it is absorbed into the systemic circulation. Peak plasma concentrations (Tmax) were observed within approximately 10.5 hours in the Phase 1 study.[32]
  • Distribution: As a GalNAc-conjugated siRNA, Lepodisiran is specifically designed for targeted delivery to the liver. The GalNAc ligands facilitate binding to the ASGPR on hepatocytes, leading to rapid and efficient receptor-mediated endocytosis into these cells.[13] This targeted uptake results in high concentrations of the siRNA in the liver, its intended site of action, while minimizing exposure to other tissues. Consistent with this liver-centric distribution, plasma levels of Lepodisiran were found to be transient, becoming undetectable by 48 hours after a single subcutaneous dose in the Phase 1 trial.[32]
  • Metabolism: Like other oligonucleotide therapeutics, siRNAs, including Lepodisiran, are expected to be metabolized by cellular endo- and exonucleases. These enzymes break down the RNA molecule into smaller, inactive nucleotide fragments.[17] The chemical modifications incorporated into the GalXC™ platform siRNAs are designed to enhance resistance to nuclease degradation, thereby prolonging intracellular stability and duration of action.[16] Within the endosomal pathway of hepatocytes, the GalNAc-siRNA conjugate is processed, leading to the release of the siRNA into the cytoplasm where it can engage with the RISC machinery.[17]
  • Excretion: The metabolic byproducts (smaller nucleotide fragments) of siRNA degradation are typically eliminated from the body primarily through renal excretion.[17] Specific excretion pathway details for Lepodisiran itself are not extensively provided in the available materials but would generally follow this pattern for GalNAc-siRNA therapeutics.

B. Plasma Half-life, Cmax, Tmax, AUC

Pharmacokinetic parameters from the Phase 1 single ascending-dose study (NCT04914546) provide initial insights into Lepodisiran's behavior in humans [32]:

  • Tmax (Time to Maximum Plasma Concentration): Approximately 10.5 hours post-subcutaneous administration.
  • Plasma Clearance: Lepodisiran was rapidly cleared from the plasma, with concentrations falling below the limit of detection by 48 hours after dosing. Specific dose-normalized Cmax (maximum plasma concentration) and AUC (area under the plasma concentration-time curve) values for each dose cohort in Phase 1 are not detailed in the summarized snippets, but these would have been key parameters evaluated to understand dose-proportionality and overall systemic exposure. A dedicated Phase 1 study in participants with varying degrees of hepatic impairment (NCT06916078 / NCT0591635) is currently underway or planned. This study will specifically assess pharmacokinetic parameters such as AUC and Cmax in these special populations to determine if dose adjustments are necessary.[15] The pharmacokinetic profile of Lepodisiran, characterized by rapid absorption to Tmax followed by swift clearance from the plasma, is consistent with the profiles of other GalNAc-conjugated siRNA therapeutics. This pattern supports the intended mechanism of rapid and efficient uptake by the liver, where the drug is sequestered and can exert its prolonged pharmacodynamic effect.

C. Relationship Between Dose/Exposure and Lp(a) Reduction (Pharmacodynamics)

A clear and consistent relationship between the administered dose of Lepodisiran and the magnitude of Lp(a) reduction has been observed across its clinical development program:

  • Phase 1 (NCT04914546): Single subcutaneous doses of Lepodisiran resulted in dose-dependent reductions in serum Lp(a) concentrations. Maximal median changes from baseline ranged from -41% with the 4 mg dose to a profound -97% with the 608 mg dose.[32]
  • Phase 2 (ALPACA Trial, NCT05565742): This multi-dose study further confirmed the dose-response relationship. The primary endpoint (placebo-adjusted time-averaged Lp(a) reduction from Day 60 to Day 180) showed reductions of -40.8% for the 16 mg dose, -75.2% for the 96 mg dose, and -93.9% for the pooled 400 mg baseline dose group.[4] The remarkable and prolonged pharmacodynamic effect (i.e., sustained suppression of Lp(a) levels for many months) despite the short plasma presence of Lepodisiran is a key characteristic. This indicates highly efficient and sustained target engagement (silencing of apo(a) mRNA) within the hepatocytes. This is likely attributable to the high stability of the siRNA molecule once it is loaded into the RISC complex within these long-lived liver cells.[17] The profound Lp(a) reduction seen at the higher doses (e.g., 400 mg and 608 mg) suggests that near-maximal target engagement is being achieved. The duration of this effect appears to be limited more by the natural turnover rate of the siRNA-RISC complex or the regeneration of hepatocytes, rather than by the systemic clearance rate of the drug from the body.

Table 7: Key Pharmacokinetic and Pharmacodynamic Parameters of Lepodisiran (from available clinical data)

ParameterValueStudy PhaseReferences
Administration RouteSubcutaneous (SC)Phase 1, 227
Tmax (Time to Peak Plasma Concentration)~10.5 hoursPhase 132
Time to Undetectable Plasma LevelsBy 48 hours post-single dosePhase 132
Lp(a) Reduction (608 mg single dose, median % change at 48 weeks)-94%Phase 125
Lp(a) Reduction (400 mg pooled, placebo-adj. time-avg % change D60-180)-93.9%Phase 24
Lp(a) Reduction (400mg x2 doses, % from baseline at Day 540)-74.2%Phase 24

IX. Regulatory Landscape and Current Development Status

A. Current Investigational Status

Lepodisiran (DCR-CM2/LY3819469) is an investigational drug currently in late-stage clinical development. It has progressed to Phase 3 clinical trials.[2] The pivotal ACCLAIM-Lp(a) cardiovascular outcomes trial (NCT06292013) is actively enrolling participants.[41]

B. Regulatory Interactions or Designations (FDA/EMA)

The available research materials do not indicate that Lepodisiran has received any specific expedited regulatory designations from major health authorities like the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA), such as Fast Track, Breakthrough Therapy, or PRIME (Priority Medicines) status.7 While Dicerna Pharmaceuticals, the originator company, received Breakthrough Therapy Designation from the FDA for a different GalXC™ platform drug, DCR-PHXC, for Primary Hyperoxaluria Type 1 14, this designation does not apply to Lepodisiran. Eli Lilly and Company has highlighted the initiation of the Phase 3 program for Lepodisiran in its investor communications, based on positive early-phase data.21

The absence of announced special regulatory designations for Lepodisiran, despite the strong Phase 2 efficacy demonstrated in Lp(a) reduction, may be attributed to the current regulatory stance on Lp(a) as a therapeutic target. Lp(a) reduction is still largely considered a surrogate endpoint by regulatory agencies. These agencies typically require robust evidence from large-scale cardiovascular outcome trials (CVOTs) demonstrating a reduction in clinically significant endpoints, such as MACE, before granting designations like Breakthrough Therapy for cardiovascular drugs. This is particularly true when the therapy targets a novel risk factor for which the direct clinical benefit of modification has not yet been unequivocally established through prospective, randomized controlled trials. The ongoing ACCLAIM-Lp(a) Phase 3 trial is specifically designed to provide this crucial outcomes data, which will be pivotal for future regulatory submissions and potential approvals.

X. Therapeutic Potential and Unmet Medical Need

A. Significance of Lp(a) as a Cardiovascular Risk Factor

Lipoprotein(a) is a well-established, independent, and predominantly genetically determined risk factor for a broad spectrum of atherosclerotic cardiovascular diseases.[1] Elevated Lp(a) levels are highly prevalent, affecting approximately one in four to one in five individuals globally.[4] Its atherogenic and thrombogenic potential contributes to an increased risk of myocardial infarction, ischemic stroke, peripheral arterial disease, and calcific aortic valve stenosis, largely independent of other traditional risk factors like LDL-cholesterol.[9] Importantly, Lp(a) concentrations are primarily set by an individual's genetic makeup (specifically, polymorphisms in the LPA gene) and remain relatively stable throughout life, largely unaffected by lifestyle modifications or most existing lipid-lowering medications.[4]

B. Limitations of Current Therapies in Lowering Lp(a)

There is a significant unmet medical need for effective and specific Lp(a)-lowering therapies. Current standard-of-care treatments for dyslipidemia, such as statins and ezetimibe, have little to no impact on Lp(a) levels; in fact, statins may even slightly increase Lp(a) concentrations in some individuals.[4] Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors can reduce Lp(a) by approximately 20-30%, but this effect is often insufficient for individuals with very high baseline levels, and these agents are primarily indicated for LDL-C reduction.[11] Niacin can lower Lp(a) to a greater extent, but its use is limited by tolerability issues (e.g., flushing) and a lack of consistent evidence for cardiovascular benefit when added to statin therapy. Lipoprotein apheresis is an effective method for acutely lowering Lp(a) but is invasive, expensive, time-consuming, and not widely accessible, typically reserved for very high-risk patients with extremely elevated Lp(a) and progressive ASCVD.[49] Consequently, millions of individuals with high Lp(a)-mediated cardiovascular risk currently lack a targeted pharmacological treatment option.

C. Potential Impact of Lepodisiran in Addressing This Unmet Need

Lepodisiran, with its ability to potently and durably reduce plasma Lp(a) concentrations by directly inhibiting apo(a) synthesis, holds the potential to be a first-in-class or best-in-class therapeutic specifically addressing this unmet medical need.1 If the ongoing ACCLAIM-Lp(a) Phase 3 cardiovascular outcomes trial successfully demonstrates that Lepodisiran-mediated Lp(a) reduction translates into a clinically meaningful decrease in MACE, it could revolutionize the management of cardiovascular risk for individuals with elevated Lp(a).4

The advent of effective and specific Lp(a)-lowering therapies like Lepodisiran could herald a new era of more personalized cardiovascular prevention. Current risk assessment and treatment paradigms are heavily centered on LDL-cholesterol. The availability of a proven therapy for elevated Lp(a) would likely lead to wider adoption of Lp(a) screening in clinical practice and enable targeted intervention for a substantial segment of the population whose cardiovascular risk remains high despite optimal management of other risk factors. This would represent a significant step towards addressing residual cardiovascular risk more comprehensively.

Furthermore, the pharmacokinetic profile of Lepodisiran, allowing for a potentially biannual or even less frequent subcutaneous injection schedule, offers a considerable advantage in terms of patient adherence. Poor adherence to daily medications is a major challenge in the long-term management of chronic conditions. An infrequent, long-acting preventative therapy could substantially improve real-world effectiveness and patient quality of life, which is critical for therapies intended for lifelong administration to mitigate cardiovascular risk.

XI. Future Directions and Conclusion

A. Summary of Key Findings

Lepodisiran (DCR-CM2/LY3819469) is an investigational GalNAc-conjugated siRNA therapeutic that acts by specifically inhibiting the hepatic synthesis of apolipoprotein(a), the defining protein component of Lp(a). Clinical studies to date (Phase 1 and Phase 2 ALPACA trial) have demonstrated that Lepodisiran can achieve profound, dose-dependent, and exceptionally durable reductions in plasma Lp(a) concentrations, with decreases exceeding 90% at optimal doses and effects lasting for up to 1.5 years with a two-dose regimen. The safety and tolerability profile observed thus far has been generally acceptable, with injection site reactions and transient liver enzyme elevations being the most notable AEs, consistent with the GalNAc-siRNA class. No drug-related serious adverse events or discontinuations due to AEs in Lepodisiran arms were reported in Phase 2.

B. Outlook Based on Ongoing Phase 3 Trial (ACCLAIM-Lp(a))

The future of Lepodisiran hinges on the outcomes of the large-scale, ongoing ACCLAIM-Lp(a) Phase 3 cardiovascular outcomes trial (NCT06292013). This pivotal study, expected to report results around 2029 [2], will determine whether the substantial Lp(a) reduction achieved with Lepodisiran translates into a statistically significant and clinically meaningful reduction in Major Adverse Cardiovascular Events (MACE) in a high-risk population with elevated Lp(a). Positive results from ACCLAIM-Lp(a) would be transformative, likely leading to regulatory approvals and establishing Lp(a) lowering as a new therapeutic strategy in cardiovascular disease prevention. Conversely, a neutral or negative outcome would raise significant questions about the clinical utility of targeting Lp(a) with this agent, despite its potent biomarker-modifying effects.

C. Concluding Remarks on the Promise of DCR-CM2 (Lepodisiran)

Lepodisiran represents a highly promising and targeted therapeutic approach to address the significant unmet medical need posed by elevated Lp(a), a prevalent and genetically determined cardiovascular risk factor for which no specific approved pharmacological treatments currently exist. Its development underscores the maturation and potential of RNA interference therapeutics, particularly when combined with advanced liver-targeting delivery platforms like GalXC™, to tackle genetically driven diseases with precision and a favorable dosing profile.

Should the Phase 3 ACCLAIM-Lp(a) trial yield positive results, Lepodisiran could not only become a valuable new medicine but also serve to definitively validate Lp(a) as a modifiable therapeutic target, potentially on par with LDL-cholesterol. Such an outcome would likely catalyze widespread changes in cardiovascular risk assessment protocols, leading to increased Lp(a) screening, and fundamentally alter treatment guidelines to incorporate targeted Lp(a)-lowering strategies. While the competitive landscape for Lp(a)-lowering therapies is emerging, with other agents also in late-stage development 20, Lepodisiran's potential for a highly infrequent dosing regimen (e.g., biannual) could offer a significant competitive advantage in terms of patient convenience and long-term adherence, provided its efficacy and safety are robustly confirmed. The journey of Lepodisiran from a novel technological platform to a potential cornerstone in cardiovascular prevention is being closely watched by the medical and scientific community.

Works cited

  1. Lepodisiran for Hyperlipidemia Treatment - Ontosight.ai, accessed June 4, 2025, https://ontosight.ai/glossary/term/lepodisiran-for-hyperlipidemia-treatment--67a0da116c3593987a502d41
  2. Lepodisiran sodium by Eli Lilly and Co for Atherosclerosis: Likelihood of Approval, accessed June 4, 2025, https://www.pharmaceutical-technology.com/data-insights/lepodisiran-sodium-eli-lilly-and-co-atherosclerosis-likelihood-of-approval/
  3. Lepodisiran - Wikipedia, accessed June 4, 2025, https://en.wikipedia.org/wiki/Lepodisiran
  4. Lilly's lepodisiran reduced levels of genetically inherited heart disease risk factor, lipoprotein(a), by nearly 94% from baseline at the highest tested dose in adults with elevated levels, accessed June 4, 2025, https://investor.lilly.com/news-releases/news-release-details/lillys-lepodisiran-reduced-levels-genetically-inherited-heart
  5. Lilly's Lepodisiran Shows 94% Reduction in Lipoprotein(a) in Phase 2 Trial, Advancing Hope for Genetic Heart Risk Treatment - BioPharma BoardRoom, accessed June 4, 2025, https://www.biopharmaboardroom.com/news/25/3120/lillys-lepodisiran-shows-94-reduction-in-lipoproteina-in-phase-2-trial-advancing-hope-for-genetic-heart-risk-treatment.html
  6. Lepodisiran, a long-duration small interfering RNA, reduces lipoprotein(a) levels, accessed June 4, 2025, https://www.2minutemedicine.com/lepodisiran-a-long-duration-small-interfering-rna-reduces-lipoproteina-levels/
  7. Lilly's lepodisiran reduced levels of genetically inherited heart disease risk factor, lipoprotein(a), by nearly 94% from baseline at the highest tested dose in adults with elevated levels - PR Newswire, accessed June 4, 2025, https://www.prnewswire.com/news-releases/lillys-lepodisiran-reduced-levels-of-genetically-inherited-heart-disease-risk-factor-lipoproteina-by-nearly-94-from-baseline-at-the-highest-tested-dose-in-adults-with-elevated-levels-302414699.html
  8. Lepodisiran Reduced Lower Serum Lp(a) by More than 90% in Phase 2 Trial - HCPLive, accessed June 4, 2025, https://www.hcplive.com/view/lepodisiran-reduced-lower-serum-lp-a-by-more-than-90-in-phase-2-trial
  9. 2024: The Year in Cardiovascular Disease - The Year of Lipoprotein(a) – Research Advances and New Findings., accessed June 4, 2025, https://www.archivesofmedicalscience.com/2024-The-Year-in-Cardiovascular-Disease-The-Year-of-Lipoprotein-a-Research-Advances,202213,0,2.html
  10. Phase II ALPACA Trial Shows Lepodisiran Produces Significant, Sustained Lipoprotein(a) Reductions, accessed June 4, 2025, https://www.appliedclinicaltrialsonline.com/view/alpaca-trial-lepodisiran-lipoprotein
  11. NLA 2025: Novel Therapeutic Approaches Show Promise for Targeted Reduction of Lp(a) in Patients With ASCVD - Pharmacy Times, accessed June 4, 2025, https://www.pharmacytimes.com/view/nla-2025-novel-therapeutic-approaches-show-promise-for-targeted-reduction-of-lp-a-in-patients-with-ascvd
  12. Lepodisiran Reduces Lipoprotein(a) by Nearly 94% in Adults With Elevated Levels of Lp(a), accessed June 4, 2025, https://www.pharmacytimes.com/view/lepodisiran-reduces-lipoprotein-a-by-nearly-94-in-adults-with-elevated-levels-of-lp-a-
  13. dicernacorporatepresenta - SEC.gov, accessed June 4, 2025, https://www.sec.gov/Archives/edgar/data/1399529/000139952920000020/dicernacorporatepresenta.htm
  14. Dicerna Receives Breakthrough Therapy Designation for DCR-PHXC for Treatment of Primary Hyperoxaluria Type 1 (PH1) - BioSpace, accessed June 4, 2025, https://www.biospace.com/dicerna-receives-breakthrough-therapy-designation-for-dcr-phxc-for-treatment-of-primary-hyperoxaluria-type-1-ph1
  15. Lepodisiran sodium - Drug Targets, Indications, Patents - Patsnap Synapse, accessed June 4, 2025, https://synapse.patsnap.com/drug/483e8f06ce7e4a94b608babcfeeb2cc1
  16. Recent Update on siRNA Therapeutics - PMC - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12026779/
  17. Whole-Body Physiologically Based Pharmacokinetic Modeling of ..., accessed June 4, 2025, https://www.mdpi.com/1999-4923/17/1/69
  18. Preclinical Toxicological Assessment of A Novel siRNA, SLN360, Targeting Elevated Lipoprotein (a) in Cardiovascular Disease - Oxford Academic, accessed June 4, 2025, https://academic.oup.com/toxsci/article/189/2/237/6613944
  19. United States Adopted Name (USAN) Drug Finder, accessed June 4, 2025, https://searchusan.ama-assn.org/finder/usan/search/LM-86/relevant/1/
  20. Lilly links lepodisiran to durably lower lipoprotein in phase 2, providing edge over Amgen and Novartis - Fierce Biotech, accessed June 4, 2025, https://www.fiercebiotech.com/biotech/lilly-links-lepodisiran-durably-lower-lipoprotein-phase-2-providing-edge-over-amgen-and
  21. Lilly reports first-quarter 2025 financial results and highlights pipeline momentum, accessed June 4, 2025, https://investor.lilly.com/news-releases/news-release-details/lilly-reports-first-quarter-2025-financial-results-and
  22. Lilly's Heart Disease Drug Shows 94% Reduction in Key Risk Factor | LLY Stock News, accessed June 4, 2025, https://www.stocktitan.net/news/LLY/lilly-s-lepodisiran-reduced-levels-of-genetically-inherited-heart-yg62etykq2sh.html
  23. Dicerna's GalXC-Plus™ RNAi Technology Delivers Target Knockdown Across CNS and to Specific CNS Cell Types in Preclinical Studies - Business Wire, accessed June 4, 2025, https://www.businesswire.com/news/home/20210330005357/en/Dicernas-GalXC-Plus-RNAi-Technology-Delivers-Target-Knockdown-Across-CNS-and-to-Specific-CNS-Cell-Types-in-Preclinical-Studies
  24. Lepodisiran - A Long-Duration Small Interfering RNA Targeting ..., accessed June 4, 2025, https://pubmed.ncbi.nlm.nih.gov/40162643/
  25. Phase 1 Trial Shows Encouraging Data for Lepodisiran as Lp(a ..., accessed June 4, 2025, https://www.physiciansweekly.com/phase-1-trial-shows-encouraging-data-for-lepodisiran-as-lpa-lowering-therapy/
  26. ACC.25 Conference Highlights: Breakthroughs in Cardiology - DelveInsight, accessed June 4, 2025, https://www.delveinsight.com/blog/acc25-conference-highlights
  27. A Study of Lepodisiran in Participants With Elevated Lipoprotein(a ..., accessed June 4, 2025, https://www.acc.org/Latest-in-Cardiology/Clinical-Trials/2025/03/27/18/03/alpaca
  28. Eli Lilly's lepodisiran cuts lipoprotein(a) by 93.9% in Phase II trial - Clinical Trials Arena, accessed June 4, 2025, https://www.clinicaltrialsarena.com/news/eli-lillys-lepodisiran-cuts-lipoproteina-by-93-9-in-phase-ii-trial/
  29. Lepodisiran — A Long-Duration Small Interfering RNA Targeting Lipoprotein(a), accessed June 4, 2025, https://www.researchgate.net/publication/390320365_Lepodisiran_-_A_Long-Duration_Small_Interfering_RNA_Targeting_Lipoproteina
  30. ALPACA: Single Injection of Novel Lepodisiran Reduces Lp(a) Concentration, accessed June 4, 2025, https://www.acc.org/Latest-in-Cardiology/Journal-Scans/2025/03/24/16/30/sun-130pm-alpaca-acc-2025
  31. Lepodisiran - Society for Vascular Medicine, accessed June 4, 2025, https://www.vascularmed.org/wp-content/uploads/2025/05/ALPACA-Trial.pdf
  32. Xiaosu Ma's research works | Eli Lilly and Company and other places - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/scientific-contributions/Xiaosu-Ma-2096960369
  33. Lepodisiran, ALPACA, and Lp(a), with Steve Nissen, MD - HCPLive, accessed June 4, 2025, https://www.hcplive.com/view/lepodisiran-alpaca-and-lp-a-with-steve-nissen-md
  34. Dicerna Prioritizes Resources to Advance GalXC™ Product Candidates, accessed June 4, 2025, https://www.americanpharmaceuticalreview.com/1315-News/191865-Dicerna-Prioritizes-Resources-to-Advance-GalXC-Product-Candidates/
  35. Pre-clinical assessment of SLN360, a novel siRNA targeting LPA, developed to address elevated lipoprotein (a) in cardiovascular disease | Request PDF - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/359675853_Pre-clinical_assessment_of_SLN360_a_novel_siRNA_targeting_LPA_developed_to_address_elevated_lipoprotein_a_in_cardiovascular_disease
  36. accessed January 1, 1970, httpsA://www.nejm.org/doi/full/10.1056/NEJMoa2415818
  37. www.nejm.org, accessed June 4, 2025, https://www.nejm.org/doi/full/10.1056/NEJMoa2415818
  38. clinicaltrials.gov, accessed June 4, 2025, https://clinicaltrials.gov/study/NCT05565742
  39. Lepodisiran Reduced Lp(a) Levels by Nearly 94% in Phase 2 ALPACA Trial, Slated for Phase 3 Development, accessed June 4, 2025, https://www.patientcareonline.com/view/lepodisiran-reduced-lp-a-levels-by-nearly-94-in-phase-2-alpaca-trial-slated-for-phase-3-development
  40. Eli Lilly's Lepodisiran Shows 95% Reduction in Lipoprotein(a), Offering New Hope for South Asian Heart Patients - MedPath, accessed June 4, 2025, https://trial.medpath.com/news/46b884df43b7d885/eli-lilly-s-lepodisiran-shows-95-reduction-in-lipoprotein-a-offering-new-hope-for-south-asian-heart-patients
  41. Lepodisiran on the Reduction of Major Adverse Cardiovascular Events in Adults With Elevated Lipoprotein(a) - ACCLAIM-Lp(a) - Clinical Trials at UCSD, accessed June 4, 2025, https://clinicaltrials.ucsd.edu/trial/NCT06292013
  42. A Study to Investigate the Effect of Lepodisiran on the Reduction of Major Adverse Cardiovascular Events in Adults With Elevated Lipoprotein(a) - ACCLAIM-Lp(a) - Stanford Health Care, accessed June 4, 2025, https://stanfordhealthcare.org/trials/a/NCT06292013.html
  43. 2025 Proxy Statement - Eli Lilly Investors, accessed June 4, 2025, https://investor.lilly.com/static-files/17ef82ba-c9fb-482d-a5e1-81ee1f25c634
  44. A Study to Investigate the Effect of Lepodisiran on the Reduction of Major Adverse Cardiovascular Events in Adults With Elevated Lipoprotein(a) - ACCLAIM-Lp(a) | TrialScreen, accessed June 4, 2025, https://app.trialscreen.org/trials/phase-3-to-investigate-effect-lepodisiran-on-reduction-major-adverse-cardiovascular-trial-nct06292013
  45. Key Clinical Trial Insights from ACC 2025, with Deepak Bhatt, MD, MPH, MBA​ - HCPLive, accessed June 4, 2025, https://www.hcplive.com/view/key-clinical-trial-insights-from-acc-2025-with-deepak-bhatt-md-mph-mba-
  46. Generalizability of REDUCE-IT eligibility criteria in a large diabetes cardiovascular outcomes trial - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10293663/
  47. A Study of Lepodisiran (LY3819469) in Participants With Normal, Mild, Moderate, or Severe Liver Function - Lilly Trials, accessed June 4, 2025, https://trials.lilly.com/en-US/trial/591635
  48. A Study of Lepodisiran (LY3819469) in Participants With Normal, Mild, Moderate, or Severe Liver Function - MedPath Clinical Trial Search, accessed June 4, 2025, https://trial.medpath.com/clinical-trial/c27e4642ff06ab4c/nct06916078-lepodisiran-hepatic-impairment-study
  49. Current and Emerging Treatment Options for Elevated Levels of Lipoprotein(a), accessed June 4, 2025, https://cme.tarsusmedicaleducation.com/sites/default/files/course/2024-06/News%20Digest%20Current%20and%20Emerging%20Treatment%20Options%20060724%20%281%29.pdf
  50. Clinical Research Update – March 2025 - Lambda CRO, accessed June 4, 2025, https://www.lambda-cro.com/clinical-research-update-march-2025/
  51. Global New Drug Research and Development Progress Weekly Report(7.1-7.7), accessed June 4, 2025, https://synapse.patsnap.com/blog/global-new-drug-research-and-development-progress-weekly-report71-77
  52. May 1, 2025 For release: Immediately Refer to: Ashley Hennessey; gentry_ashley_jo@lilly.com; (317) 416-4363 (Media) Mike Czapar;, accessed June 4, 2025, https://investor.lilly.com/static-files/939286fe-0214-4f10-8f41-424ca3d54840
  53. ELI LILLY AND COMPANY, accessed June 4, 2025, https://investor.lilly.com/static-files/ff06ed54-9f64-4b7f-8529-c0278d635eac
  54. Form 10-Q for ELI Lilly Co filed 08/08/2024, accessed June 4, 2025, https://investor.lilly.com/static-files/7ae2646d-3408-43e4-a9c8-18762fa18ef8
  55. Phase III HERCULES Trial Shows Tolebrutinib Significantly Slows Disability Progression in Non-Relapsing Secondary Progressive MS - Applied Clinical Trials, accessed June 4, 2025, https://www.appliedclinicaltrialsonline.com/view/hercules-trial-tolebrutinib-disability-secondary-progressive-ms?utm_source=www.appliedclinicaltrialsonline.com&utm_medium=viewNext
  56. Form 10-K for ELI Lilly Co filed 02/21/2024, accessed June 4, 2025, https://investor.lilly.com/static-files/9a5d6b82-282a-4a1c-b715-410a9059f69b
  57. Advances in nucleic acid-targeted therapies for cardiovascular disease prevention - Oxford Academic, accessed June 4, 2025, https://academic.oup.com/cardiovascres/article/120/10/1107/7708695
  58. bluebird bio Receives FDA Accelerated Approval for SKYSONA, accessed June 4, 2025, https://pipelinereview.com/index.php/2022091781638/DNA-RNA-and-Cells/bluebird-bio-Receives-FDA-Accelerated-Approval-for-SKYSONA-Gene-Therapy-for-Early-Active-Cerebral-Adrenoleukodystrophy-CALD.html

Published at: June 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.