Emerging Lp(a) Therapies: The Drug Development Pipeline
MEDICAL DISCLAIMER
Always consult a licensed healthcare professional when deciding on medical care. The information presented on this website is for educational purposes only and exclusively intended to help consumers understand the different options offered by healthcare providers to prevent, diagnose, and treat health conditions. It is not a substitute for professional medical advice when making healthcare decisions.
Introduction
After decades of recognizing Lp(a) as a cardiovascular risk factor without effective treatments, the therapeutic landscape is transforming. Multiple drugs targeting apolipoprotein(a) production are now in late-stage clinical development. These RNA-targeted therapies achieve Lp(a) reductions of 80-95%, levels that could finally validate whether lowering Lp(a) prevents cardiovascular events.
This article examines the leading candidates, their mechanisms, clinical trial data, and realistic timelines for approval. Understanding the pipeline helps patients with elevated Lp(a) plan for a future where targeted treatment becomes available, whether through clinical trial participation or eventual commercial access.
What is pelacarsen and how does it work?
Pelacarsen (formerly AKCEA-APO(a)-LRx or TQJ230) is an antisense oligonucleotide (ASO) that reduces Lp(a) production in the liver. ASOs are short, synthetic strands of nucleotides designed to bind to specific messenger RNA sequences. When pelacarsen binds to apolipoprotein(a) mRNA in hepatocytes, it triggers degradation of the mRNA, preventing translation into apo(a) protein.
With less apo(a) produced, fewer Lp(a) particles are assembled and secreted into the bloodstream. In Phase 2 trials, pelacarsen demonstrated dose-dependent Lp(a) reductions up to approximately 80% (Tsimikas et al., 2020). The highest dose (60 mg monthly) produced the greatest effect while maintaining acceptable tolerability.
Pelacarsen is developed by Ionis Pharmaceuticals in partnership with Novartis. The drug is administered by subcutaneous injection, initially monthly in the ongoing Phase 3 trial. The most common side effects in earlier studies were injection site reactions, which were generally mild.
What is the HORIZON outcomes trial?
HORIZON is the pivotal Phase 3 cardiovascular outcomes trial testing whether pelacarsen reduces major adverse cardiovascular events (MACE) in patients with established cardiovascular disease and elevated Lp(a). The trial enrolled approximately 8,000 patients with Lp(a) ≥70 mg/dL and a history of MI, stroke, or symptomatic peripheral artery disease.
Participants were randomized to pelacarsen 80 mg subcutaneously once monthly or placebo, with the primary endpoint being a composite of cardiovascular death, non-fatal MI, non-fatal stroke, and urgent coronary revascularization. The trial design reflects current regulatory expectations for cardiovascular drug approval (Tsimikas et al., 2021).
HORIZON results are anticipated in late 2025 or 2026, depending on event accrual rates. A positive result would represent the first validation of the Lp(a) hypothesis—that lowering Lp(a) reduces cardiovascular events—and would likely lead to FDA approval for pelacarsen.
What is olpasiran and how does it differ from pelacarsen?
Olpasiran (AMG 890) is a small interfering RNA (siRNA) developed by Amgen that also targets apolipoprotein(a) mRNA. Like pelacarsen, it reduces apo(a) production in the liver. The key difference is the molecular platform: siRNA works through a different cellular mechanism than ASO, leading to potentially different pharmacokinetic profiles.
In Phase 2 trials, olpasiran demonstrated remarkably potent Lp(a) lowering. The highest doses achieved greater than 95% reductions in Lp(a) that were sustained for months after each injection (Tsimikas et al., 2021). Some participants had Lp(a) levels reduced to nearly undetectable.
The duration of effect is a key practical advantage. While pelacarsen requires monthly injections, olpasiran’s effects persist for 3-6 months after each dose. Less frequent dosing could improve adherence and convenience, though ultimately clinical outcomes matter more than dosing schedules.
What is the OCEAN(a) outcomes trial?
OCEAN(a) is Amgen’s Phase 3 cardiovascular outcomes trial for olpasiran. Like HORIZON, it enrolls patients with established atherosclerotic cardiovascular disease and elevated Lp(a) (≥200 nmol/L, approximately 80 mg/dL). Approximately 6,000 patients are randomized to olpasiran or placebo.
The primary endpoint is a composite of major cardiovascular events including cardiovascular death, MI, stroke, and urgent coronary revascularization. Olpasiran is administered quarterly (every 12 weeks) via subcutaneous injection. The trial is designed to demonstrate that the profound Lp(a) reductions observed in Phase 2 translate to clinical benefit.
OCEAN(a) results are expected around 2026-2027. If both HORIZON and OCEAN(a) succeed, the Lp(a) treatment landscape will be transformed, with two different therapeutic approaches (ASO and siRNA) both proven effective. If one succeeds and the other fails, the differences in platforms and dosing may provide mechanistic insights.
Discover the tests and treatments that could save your life
Get our unbiased and comprehensive report on the latest techniques for heart disease prevention, diagnosis, and treatment.
What other siRNA candidates are in development?
Beyond olpasiran, several other companies are developing siRNA therapies targeting Lp(a). SLN360 (Silence Therapeutics) has completed Phase 1 trials showing substantial Lp(a) lowering. LY3819469 (Eli Lilly) is another siRNA candidate in early development. Lepodisiran (LIB003, Li Ka Shing/Arrowhead) has shown promising Phase 2 results.
Competition in this space is intense because the market opportunity for effective Lp(a)-lowering therapy is substantial. With roughly one billion people globally estimated to have elevated Lp(a), and growing recognition of Lp(a) as an undertreated risk factor, multiple pharmaceutical companies are investing heavily.
For patients, this competition is beneficial. Multiple approved therapies would provide options if one has tolerability issues, create price competition, and accelerate understanding of the Lp(a) hypothesis through diverse clinical data.
Are there oral small molecule approaches?
The development of oral small molecules targeting Lp(a) would be attractive from a convenience standpoint. However, Lp(a) biology presents challenges. The LPA gene’s complex structure (variable kringle repeats) makes traditional small molecule drug design difficult. No oral Lp(a)-lowering drugs are currently in advanced clinical development.
Some existing oral medications have been explored for Lp(a) effects. Mipomersen, an oral ASO targeting apoB, reduces Lp(a) modestly but has limiting hepatic toxicity. Lomitapide (Juxtapid), approved for homozygous familial hypercholesterolemia, affects Lp(a) minimally. Neither represents a practical Lp(a)-focused approach.
Injectable RNA-targeted therapies remain the most advanced and promising approach. Given the infrequent dosing possible with siRNA (quarterly or less frequent), the convenience gap between injectable and oral administration may be acceptable if clinical benefits are proven.
What about gene editing approaches?
CRISPR and related gene editing technologies offer the theoretical possibility of permanently reducing Lp(a) through one-time treatment. Early-stage research has demonstrated that editing the LPA gene in animal models can eliminate Lp(a) production. Verve Therapeutics is developing liver-targeted gene editing approaches for cardiovascular conditions, though Lp(a) is not their initial target.
Gene editing for Lp(a) faces significant hurdles. Safety concerns about off-target effects, immune responses, and long-term consequences of permanent genetic changes require extensive investigation. Regulatory pathways for one-time gene editing therapies remain uncertain, particularly for common conditions like elevated Lp(a).
The timeline for gene editing approaches to Lp(a) likely extends well beyond the next decade. RNA-targeted therapies that temporarily reduce apo(a) production represent a more proximate solution, with the advantage of reversibility if unexpected problems arise.
What endpoints will FDA require for approval?
The FDA has historically required demonstration of clinical outcomes (reduced cardiovascular events) for approval of lipid-lowering therapies intended for risk reduction. This contrasts with the surrogate endpoint approach (LDL cholesterol reduction) that enabled earlier statin approvals. For Lp(a) therapies, the regulatory bar is cardiovascular outcomes.
Accelerated approval based on Lp(a) reduction as a surrogate endpoint is theoretically possible but would be unprecedented. The FDA has indicated openness to discussing whether Lp(a) could qualify as a reasonably likely surrogate, but no formal determination has been made (Tsimikas et al., 2021). Companies have chosen to pursue outcomes trials rather than rely on surrogate pathway uncertainty.
If outcomes trials succeed, approval should follow relatively quickly given the established regulatory framework for cardiovascular drugs. If outcomes trials fail despite substantial Lp(a) lowering, the field would face fundamental questions about the causal role of Lp(a) in human disease.
Discover the tests and treatments that could save your life
Get our unbiased and comprehensive report on the latest techniques for heart disease prevention, diagnosis, and treatment.
What is a realistic timeline for first approval?
Assuming HORIZON or OCEAN(a) meet their primary endpoints, FDA approval could occur in 2026-2027. The timeline depends on trial completion dates, FDA review priorities, and whether any safety signals emerge requiring additional analysis.
Following FDA approval, commercial launch would occur within months. Initial access might be limited by manufacturing capacity, payer coverage decisions, and healthcare provider education. High-risk patients with documented elevated Lp(a) and established cardiovascular disease would likely receive priority access.
Broader adoption would evolve over years as guidelines incorporate the new therapies, more physicians become comfortable prescribing them, and insurance coverage expands. The path from approval to widespread use typically takes 3-5 years for cardiovascular drugs, though the trajectory could be faster given the unmet need for Lp(a) treatment.
Will approval be for Lp(a) lowering or ASCVD risk reduction?
The likely FDA approval indication would be for reducing cardiovascular risk in patients with elevated Lp(a) and established atherosclerotic cardiovascular disease, similar to how evolocumab and alirocumab are indicated for reducing cardiovascular events in patients with established disease.
Whether approval extends to primary prevention (patients without established disease) would depend on trial designs and FDA interpretation. Neither HORIZON nor OCEAN(a) enrolls primary prevention patients, so initial approval will be for secondary prevention only. Extensions to primary prevention might require additional trials or label expansion based on accumulated evidence.
The specific indication language matters for insurance coverage. Narrow indications restrict reimbursement to specific populations, while broader language enables wider use. Advocacy efforts and real-world evidence after initial approval often drive eventual expansion of approved indications.
How should patients interpret Phase 2 data?
Phase 2 trials demonstrate that a therapy can lower Lp(a) substantially, but they don’t prove clinical benefit. The dose-dependent reductions seen with pelacarsen (Tsimikas et al., 2020) and the profound reductions seen with olpasiran are necessary but not sufficient evidence that these drugs will prevent heart attacks and strokes.
Patients should interpret Phase 2 data as reason for cautious optimism rather than certainty. Many drugs that successfully lower surrogate markers have failed to improve outcomes in Phase 3 trials. Niacin’s failure despite Lp(a) lowering serves as a cautionary example, though the mechanistic situation for RNA-targeted therapies is likely different.
For clinical trial participation decisions, Phase 2 data inform the probability of eventual benefit. Strong biomarker effects suggest the therapy might work, which makes trial participation more attractive. But placebo-controlled trials always involve uncertainty about individual outcomes.
Conclusion
The Lp(a) therapeutic pipeline represents the most significant advance in cardiovascular prevention since PCSK9 inhibitors. Pelacarsen and olpasiran both achieve Lp(a) reductions comparable to what statins achieve for LDL cholesterol. If outcomes trials validate these approaches, patients with elevated Lp(a) will finally have effective targeted treatment.
The next few years will be decisive. HORIZON and OCEAN(a) results will either confirm decades of genetic and epidemiological evidence or force reconsideration of the Lp(a) hypothesis. For patients with elevated Lp(a), this period offers both uncertainty and opportunity.
Clinical trial participation provides early access to potentially transformative therapies. For those not in trials, optimizing all current treatment options and modifiable risk factors remains the practical approach while awaiting outcomes data and eventual drug approvals.
