Dalcetrapib (DrugBank ID: DB12181) is an investigational small molecule drug that represents one of the most compelling and complex narratives in modern cardiovascular pharmacology. Initially developed as a broad-application therapy to reduce cardiovascular risk by raising high-density lipoprotein cholesterol (HDL-C), its journey has been marked by a high-profile clinical trial failure, subsequent scientific redemption through a landmark pharmacogenomic discovery, and a pioneering pivot toward precision medicine. As a modulator of Cholesteryl Ester Transfer Protein (CETP), Dalcetrapib was designed to inhibit the transfer of cholesteryl esters from anti-atherogenic HDL to pro-atherogenic lipoproteins. This mechanism reliably produced a significant 30-40% increase in HDL-C levels. However, unlike other potent CETP inhibitors, it had a negligible effect on low-density lipoprotein cholesterol (LDL-C), making it an almost pure pharmacological test of the "HDL hypothesis"—the long-held belief that raising HDL-C levels would directly translate to reduced cardiovascular events.
The definitive test of this hypothesis, the dal-OUTCOMES trial involving over 15,000 patients with recent acute coronary syndrome (ACS), was terminated in 2012 for futility. Despite successfully raising HDL-C, Dalcetrapib demonstrated no clinical benefit, a result that profoundly challenged the prevailing dogma in lipidology and contributed to a paradigm shift away from HDL-C quantity as a primary therapeutic target. The drug's development was halted, and it was considered another casualty in the difficult history of CETP inhibitors.
The story was dramatically revived by a post-hoc genome-wide association study of the dal-OUTCOMES trial participants. This analysis uncovered a powerful interaction between Dalcetrapib and a specific genetic variant (rs1967309) in the adenylate cyclase type 9 (ADCY9) gene. Patients with the AA genotype experienced a 39% reduction in cardiovascular events on the drug, whereas those with the GG genotype experienced a 27% increase in risk. This finding transformed Dalcetrapib from a failed drug into a potential first-in-class precision medicine for a genetically defined subpopulation.
Subsequent development, spearheaded by DalCor Pharmaceuticals, has focused on validating this pharmacogenomic hypothesis. The prospective dal-GenE trial, which enrolled over 6,000 post-ACS patients with the protective AA genotype, yielded complex results. While it did not meet its primary composite endpoint in the main analysis, it showed a significant 21% reduction in the key secondary endpoint of myocardial infarction and demonstrated benefit in several pre-specified sensitivity analyses. A confirmatory trial, Dal-GenE-2, is now underway to provide the definitive evidence required for potential regulatory approval. This report provides a comprehensive analysis of Dalcetrapib, detailing its chemical properties, its unique pharmacological profile, the full history of its clinical development, the scientific basis of its pharmacogenomic revival, and its current standing as a potential harbinger of a new era of genetically-guided therapy in cardiovascular medicine.
Dalcetrapib is a small molecule drug with a well-defined chemical identity, crucial for its classification and understanding its pharmacological behavior.[1]
The physicochemical properties of Dalcetrapib are fundamental to its absorption, distribution, metabolism, and excretion (ADME) profile and its mechanism of action. It is chemically classified as an anilide, belonging to the superclass of benzenoids.[1] A key structural feature is its thioester group, which defines its nature as a prodrug.[3]
Dalcetrapib is characterized by its high lipophilicity and extremely low aqueous solubility. Its predicted octanol-water partition coefficient (logP) is high, with values ranging from 6.24 to 7.92, indicating a strong preference for lipid environments over aqueous ones.[1] This is complemented by a very low water solubility of 0.000392 mg/mL, rendering it practically insoluble.[1] While poorly soluble in water, it is soluble in organic solvents such as dimethyl sulfoxide (DMSO) and ethanol.[4]
These properties result in violations of several standard rules used to predict oral bioavailability, including Lipinski's Rule of Five, the Ghose Filter, and Veber's Rule.[1] Despite these predictions suggesting potential pharmacokinetic challenges, some computational models predict good bioavailability.[1] This combination of extreme lipophilicity, poor water solubility, and its status as a thioester prodrug that requires in vivo hydrolysis to its active thiol form creates a complex ADME profile.[8] This profile likely dictates its partitioning into the lipid-rich environments of lipoproteins and cell membranes, which is central to its interaction with its target, CETP. The site and rate of its enzymatic activation could introduce significant inter-individual variability in the concentration of the active drug at the target site, a factor that may underpin the pronounced pharmacogenomic effect discovered later in its development.
| Property | Value | Source(s) | 
|---|---|---|
| DrugBank ID | DB12181 | 1 | 
| CAS Number | 211513-37-0 | 1 | 
| Chemical Formula | 2 | |
| Molecular Weight | 389.6 g/mol | 4 | 
| Type | Small Molecule, Prodrug | 1 | 
| Chemical Class | Anilide, Thioester | 1 | 
| Water Solubility | 0.000392 mg/mL | 1 | 
| logP | 6.24 - 7.92 | 1 | 
| pKa (Strongest Acidic) | 12.65 - 13.05 | 1 | 
| Hydrogen Bond Donors | 1 | 1 | 
| Hydrogen Bond Acceptors | 2 - 3 | 1 | 
| Rule of Five Violation | Yes (1 violation) | 1 | 
To comprehend the action of Dalcetrapib, it is essential to first understand its molecular target, Cholesteryl Ester Transfer Protein (CETP). CETP is a hydrophobic glycoprotein synthesized primarily in the liver and adipose tissue that circulates in the plasma, mostly bound to HDL particles.[1] Its primary function is to facilitate the transfer of neutral lipids, specifically cholesteryl esters (CE) and triglycerides (TG), among the different classes of lipoproteins.[1]
The net effect of CETP activity is the equimolar exchange of CE from HDL particles to apolipoprotein B (ApoB)-containing lipoproteins—namely very-low-density lipoproteins (VLDL) and low-density lipoproteins (LDL)—in return for TG.[1] This process is a critical node in the reverse cholesterol transport pathway, by which excess cholesterol from peripheral tissues is returned to the liver for excretion. By moving cholesterol away from the supposedly anti-atherogenic HDL to the known pro-atherogenic LDL and VLDL particles, CETP activity was considered a detrimental process in the context of atherosclerosis.[11] Consequently, the therapeutic hypothesis emerged that inhibiting CETP would be cardioprotective by trapping cholesterol within the HDL fraction, thereby raising HDL-C levels, and, in the case of more potent inhibitors, reducing the cholesterol content of LDL particles.[13]
Dalcetrapib is classified as a CETP inhibitor, or more accurately, a CETP modulator.[3] As a thioester prodrug, it undergoes in vivo hydrolysis to its active thiol form, which then interacts with CETP.[8] Evidence suggests it binds covalently to a specific cysteine residue (Cys-13) on the CETP molecule.[15]
Its mechanism is distinct from other CETP inhibitors like torcetrapib or anacetrapib. Dalcetrapib appears to be a selective modulator of CETP's function. It primarily inhibits the heterotypic transfer of lipids—the exchange between different lipoprotein classes (e.g., HDL to LDL)—but does not significantly affect homotypic transfers, which occur between particles of the same class (e.g., within the HDL subclass population).[8] This selective action suggests a more subtle modulation of the CETP pathway rather than a complete and non-specific blockade.
Consistent with this modulated mechanism, Dalcetrapib is considered a modest CETP inhibitor in terms of potency when compared to other agents in its class.[17] In clinical trials, a 600 mg daily dose of Dalcetrapib typically resulted in a 30% to 56% reduction in plasma CETP activity.[11]
The pharmacological action of Dalcetrapib translates into a distinct and consistent pattern of changes in the plasma lipid profile.
The initial clinical development of Dalcetrapib was conducted by F. Hoffmann-La Roche under a global program named dal-HEART, which comprised six clinical trials designed to establish the drug's efficacy on surrogate markers and its safety profile.[24] These foundational studies were critical in differentiating Dalcetrapib from the failed first-generation CETP inhibitor, torcetrapib, and building the case for a large-scale cardiovascular outcomes trial.
Key trials in this program included:
The collective results from these trials painted a promising picture: Dalcetrapib effectively raised HDL-C, appeared to have a favorable safety profile devoid of torcetrapib-like toxicity, and showed positive effects on surrogate markers of atherosclerosis. This body of evidence provided the rationale for proceeding with the definitive, large-scale Phase III cardiovascular outcomes trial.
The dal-OUTCOMES study was the pivotal Phase III trial designed to determine if the HDL-C raising effect of Dalcetrapib would translate into a reduction in major cardiovascular events.[34]
The dal-OUTCOMES trial successfully replicated the pharmacodynamic effects of Dalcetrapib seen in earlier studies. Over a median follow-up of 31 months, HDL-C levels increased by 31% to 40% in the Dalcetrapib group, compared to a modest 4% to 11% increase in the placebo group. As expected, Dalcetrapib had a minimal effect on LDL-C levels.[17]
Despite this robust and sustained effect on the target lipid biomarker, Dalcetrapib failed to show any clinical benefit. The trial was stopped prematurely based on the recommendation of the independent Data and Safety Monitoring Board (DSMB) following a pre-specified interim analysis.[22] The analysis revealed a clear lack of efficacy, or futility.
The final results showed that Dalcetrapib did not alter the risk of the primary endpoint. The cumulative event rate was 8.3% in the Dalcetrapib group versus 8.0% in the placebo group, yielding a hazard ratio (HR) of 1.04 (95% confidence interval [CI], 0.93 to 1.16; P=0.52).[21] Furthermore, no benefit was observed for any of the individual components of the primary endpoint, and analyses of various predefined subgroups based on baseline clinical or biochemical characteristics also failed to identify any population that derived a benefit from the treatment.[34]
In May 2012, Roche announced the termination of the dal-OUTCOMES trial and the discontinuation of the entire dal-HEART clinical development program.[28] The decision was based solely on the DSMB's finding of a lack of clinically meaningful efficacy.[8] Importantly, the DSMB reported no specific safety concerns that would warrant termination.[28]
However, the final publication of the trial results did reveal two small but statistically significant adverse signals in the Dalcetrapib group: a mean increase in systolic blood pressure of 0.6 mm Hg and a median increase in the inflammatory marker C-reactive protein (CRP) of 0.2 mg/L, when compared with placebo.[21] While minor, these directionally unfavorable changes may have contributed to offsetting any potential small benefit of the drug.
The failure of dal-OUTCOMES was a profound disappointment and a pivotal moment in cardiovascular research. It demonstrated a stark disconnect between a drug's ability to favorably modulate a surrogate biomarker (HDL-C) and its ability to improve hard clinical outcomes. This translational failure served as a powerful cautionary tale about the limitations of relying on surrogate endpoints to predict clinical success in drug development and dealt a severe blow to the simple "HDL hypothesis."
| Trial Name (NCT ID) | Phase | Patient Population | N | Primary Objective / Endpoint | Key Finding | 
|---|---|---|---|---|---|
| dal-VESSEL (part of NCT00658515) | IIb | Patients with or at risk of CHD | 476 | Change in endothelial function (FMD) and blood pressure | Increased HDL-C without adverse effects on FMD or blood pressure; demonstrated lack of torcetrapib-like toxicity.11 | 
| dal-PLAQUE | IIb | Patients with atherosclerosis | 130 | Change in atherosclerotic plaque burden via imaging | Showed a promising signal for reduced total vessel area and plaque burden at 24 months.30 | 
| dal-ACUTE (NCT01323153) | III | Patients hospitalized for ACS | 300 | Percent change in HDL-C at 4 weeks | Safe and effective at raising HDL-C when initiated early after an ACS event.27 | 
| dal-OUTCOMES (NCT00658515) | III | Stable post-ACS patients | 15,871 | Composite of major adverse cardiovascular events (MACE) | Terminated for futility; Dalcetrapib raised HDL-C by 31-40% but did not reduce the risk of MACE (HR 1.04).21 | 
| dal-GenE (NCT02525939) | III | Post-ACS patients with ADCY9 AA genotype | 6,147 | Composite of MACE | Did not meet primary endpoint (HR 0.88, P=0.12), but showed a 21% reduction in MI (HR 0.79, P=0.02).40 | 
Following the discontinuation of Dalcetrapib's development, the narrative took an unexpected and dramatic turn. In 2012, investigators at the Montreal Heart Institute conducted a retrospective, post-hoc genome-wide association study (GWAS) on DNA samples from 5,749 participants of the dal-OUTCOMES trial.[34] The analysis aimed to identify genetic factors that might have influenced the response to Dalcetrapib.
The study yielded a remarkable result: a single nucleotide polymorphism (SNP), rs1967309, located in an intron of the adenylate cyclase type 9 (ADCY9) gene on chromosome 16, showed a highly significant association with cardiovascular outcomes, but exclusively in the patients treated with Dalcetrapib.[13] There was no association between this SNP and outcomes in the placebo group.
The effect of Dalcetrapib was powerfully stratified by the patient's genotype at this specific locus:
This striking finding suggested that the overall neutral result of the dal-OUTCOMES trial was an average of three distinct effects: substantial benefit in one genetic subgroup, no effect in another, and potential harm in a third. This discovery provided a compelling hypothesis to explain the trial's failure and offered a new path forward for Dalcetrapib as a precision medicine. The rights to the drug were subsequently acquired by DalCor Pharmaceuticals, which began a new development program focused exclusively on patients with the protective AA genotype, complete with a co-developed companion diagnostic test to identify them.[6]
The discovery of the ADCY9 interaction prompted intense investigation into the gene's role in cardiovascular biology. ADCY9 encodes adenylate cyclase 9, a membrane-bound enzyme responsible for synthesizing the critical second messenger cyclic AMP (cAMP).[46] It is widely expressed, including in cardiovascular tissues such as the heart and vascular smooth muscle.[47]
Preclinical research has established a direct functional link between the ADCY9 and CETP pathways. In mouse models, inactivation of the Adcy9 gene was found to be atheroprotective, leading to a 65% reduction in atherosclerosis and improved endothelial function.[48] Crucially, this protective effect was abolished when the mice were also transgenic for human CETP, confirming a biological interaction between the two pathways.[17] Further studies have shown that Adcy9 inactivation improves cardiac function and remodeling after myocardial infarction in mice.[50]
The proposed molecular mechanism for the pharmacogenomic interaction centers on the macrophage within the arterial wall. The hypothesis posits that the protective AA genotype at rs1967309 leads to reduced ADCY9 activity or expression. This, in turn, paradoxically results in higher intracellular cAMP levels. Elevated cAMP is known to promote the expression of transporters like ABCA1, which are critical for cholesterol efflux from macrophages to HDL particles. When a patient with the AA genotype is treated with Dalcetrapib, the drug's effect is layered upon this favorable genetic background. The enhanced cholesterol efflux capacity may counteract any potentially detrimental intracellular effects of CETP modulation, leading to a net anti-atherosclerotic and clinically protective outcome.[43]
To move beyond the limitations of a post-hoc finding, the dal-GenE trial was designed as a prospective, randomized, controlled trial to definitively test the ADCY9 pharmacogenomic hypothesis.[13]
The results of the dal-GenE trial, published in 2022, were complex and nuanced, falling short of a clear, unqualified success but providing strong signals of a drug effect.[6]
The results of dal-GenE represent a landmark, though complicated, test of a pharmacogenomic hypothesis in cardiology. The failure to meet the primary endpoint in the ITT analysis prevents it from being a definitive success. However, the consistent and significant benefit seen in the hard endpoint of myocardial infarction, supported by multiple pre-specified sensitivity analyses, provides a strong signal of biological activity and clinical benefit in this genetically selected population. This ambiguity has necessitated a second, confirmatory trial to resolve the question for regulators and clinicians.
Across its extensive clinical development program, Dalcetrapib has demonstrated a generally favorable safety and tolerability profile. In multiple Phase II and Phase III trials, the incidence and types of adverse events were broadly similar between the Dalcetrapib and placebo groups.[23] The most commonly reported adverse events were typically mild to moderate and included nasopharyngitis, diarrhea, bronchitis, and back pain.[30] In the dal-GenE trial, which specifically studied the AA genotype population, diarrhea was the only adverse event of interest that was more frequently observed with Dalcetrapib compared to placebo.[16]
A critical aspect of Dalcetrapib's safety assessment was to ensure it did not share the specific off-target toxicities that led to the catastrophic failure of the first CETP inhibitor, torcetrapib. Torcetrapib was found to increase blood pressure and mortality through effects on the renin-angiotensin-aldosterone system, which were independent of its action on CETP.[11]
Dalcetrapib was rigorously evaluated for these effects and was found to be free of this class of toxicity.
The absence of these severe off-target effects demonstrated that the CETP inhibition mechanism was not inherently toxic and that the adverse outcomes seen with torcetrapib were specific to that molecule.
While Dalcetrapib was demonstrably safer than torcetrapib, the large-scale dal-OUTCOMES trial, with its substantial statistical power, did identify subtle but statistically significant adverse signals. These effects are thought to be related to the on-target mechanism of CETP modulation rather than off-target toxicity.
The story of Dalcetrapib is best understood within the context of the entire class of CETP inhibitors, which has experienced a tumultuous development history. Four major agents have progressed to large-scale Phase III cardiovascular outcomes trials, each with a distinct profile and fate.
The key distinction among these agents lies in their potency and their differential effects on HDL-C and LDL-C. Dalcetrapib was a modest CETP modulator with a clean, HDL-C-specific effect. In contrast, anacetrapib and evacetrapib were highly potent inhibitors that produced massive increases in HDL-C alongside significant, statin-like reductions in LDL-C. This difference in LDL-C lowering proved to be the critical determinant of their clinical outcomes. While Dalcetrapib and evacetrapib failed due to a lack of efficacy, anacetrapib was the only agent to demonstrate a modest but statistically significant reduction in cardiovascular events. This success is now widely attributed almost entirely to its LDL-C lowering effect, not its HDL-C raising capability.[18] This comparative analysis underscores that the magnitude and type of lipid modulation are paramount; Dalcetrapib's modest, HDL-only effect was insufficient to provide a clinical benefit in a broad population.
| Feature | Torcetrapib | Dalcetrapib | Evacetrapib | Anacetrapib | 
|---|---|---|---|---|
| Lead Company | Pfizer | Roche / DalCor | Eli Lilly | Merck | 
| Potency | Potent | Modest | Potent | Potent | 
| HDL-C Effect | ~+72% | ~+30-40% | ~+130% | ~+104% | 
| LDL-C Effect | Moderate reduction | None | ~-37% | ~-17% | 
| Key Outcomes Trial | ILLUMINATE | dal-OUTCOMES | ACCELERATE | REVEAL | 
| Primary Outcome | Increased MACE & mortality | No effect on MACE | No effect on MACE | Reduced MACE by 9% | 
| Key Safety Concern | Off-target adrenal toxicity, ↑BP | Small ↑BP & CRP | Small ↑BP | Small ↑BP, long half-life | 
| Development Status | Terminated (2006) | Investigational (Phase III for genetic subgroup) | Terminated (2015) | Development not pursued (2017) | 
The collective results from the CETP inhibitor trials have fundamentally reshaped the field of lipidology by largely dismantling the simple "HDL hypothesis." For decades, the strong inverse correlation between HDL-C levels and cardiovascular risk in observational studies led to the belief that raising HDL-C would be a potent therapeutic strategy.[14]
The unequivocal failure of Dalcetrapib in dal-OUTCOMES, a trial that cleanly tested this hypothesis by raising HDL-C without affecting LDL-C, was a critical blow.[12] The subsequent failure of the potent inhibitor evacetrapib, despite a massive 130% increase in HDL-C, further solidified this conclusion.[18] These results, combined with evidence from Mendelian randomization studies showing that genetic variants that raise HDL-C do not necessarily protect against heart disease, have led to a paradigm shift.[12]
The scientific consensus now holds that the quantity of HDL-C is not a causal factor in cardiovascular disease and is therefore not a viable therapeutic target in itself. Instead, the focus has shifted towards the functionality of HDL particles (e.g., their cholesterol efflux capacity) and has strongly reinforced the primacy of lowering the concentration of atherogenic ApoB-containing lipoproteins, principally LDL, as the central goal of lipid-modifying therapy.[10]
Dalcetrapib is not an approved medication in any jurisdiction. Searches of the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA) databases do not show any past or present marketing applications for the drug.[59] Currently, no CETP inhibitors are approved for clinical use.[61]
Dalcetrapib's current status is investigational, with its development being advanced by DalCor Pharmaceuticals exclusively for the genetically defined population of post-ACS patients with the ADCY9 rs1967309 AA genotype.[6] Following the mixed results of the dal-GenE trial, a second, confirmatory Phase III trial is now underway. This study, Dal-GenE-2 (DAL-302), is designed to provide the definitive evidence on the drug's efficacy in reducing myocardial infarction in this specific population.[6] Critically, this trial is being conducted under a Special Protocol Assessment (SPA) agreement with the FDA. An SPA is a process in which the FDA provides a sponsor with a binding agreement that the design and planned analysis of a clinical trial are adequate to support a future marketing application, should the trial meet its specified endpoints.[6] This agreement significantly de-risks the regulatory path forward for Dalcetrapib, though it does not guarantee a successful trial outcome or final approval.
The trajectory of Dalcetrapib is a microcosm of major shifts in pharmaceutical research and development over the past two decades. It began as a product of the blockbuster era, designed to test a broad, simple hypothesis in a massive, heterogeneous population. Its failure in dal-OUTCOMES was not merely the failure of a single drug but a powerful refutation of the simplistic "HDL hypothesis" that had guided lipid research for years. The program's initial termination underscored the immense risk of relying on surrogate biomarkers and the frequent disconnect between promising Phase II data and definitive Phase III outcomes.
However, the subsequent discovery of the ADCY9 pharmacogenomic interaction represents a paradigm shift toward a more nuanced, data-driven, and personalized approach to medicine. The ability to retrospectively mine genomic data from a "failed" trial to uncover a signal of profound benefit in a specific subgroup has created a new model for value creation in drug development. Dalcetrapib's story demonstrates that a drug's overall neutral effect can mask powerful, opposing effects in genetically distinct subpopulations. This has transformed the drug from a clinical and commercial write-off into a pioneering agent with the potential to fill a specific, genetically defined niche.
The future of Dalcetrapib now hinges entirely on the outcome of the Dal-GenE-2 confirmatory trial.[6] Given the mixed results of the first dal-GenE study—a missed primary endpoint but a significant benefit on the hard clinical outcome of myocardial infarction—this second trial is essential to provide the unambiguous evidence of efficacy that regulators will require. The trial's focus on the endpoint of fatal and non-fatal MI is a strategic decision based on the strongest signal from the previous study.[42]
The Special Protocol Assessment agreement with the FDA provides a clear regulatory pathway, indicating that if Dal-GenE-2 successfully meets its pre-specified endpoints, the clinical evidence base should be sufficient to support an approval decision.[6] Success in this trial would validate the entire pharmacogenomic hypothesis and establish Dalcetrapib as a viable therapeutic option for a well-defined patient population. Failure would likely mark the definitive end of its long and complex development journey.
Regardless of its ultimate fate, the Dalcetrapib saga has already left an indelible mark on cardiovascular medicine. It stands as the most advanced and visible test case for a pharmacogenomically-guided therapy aimed at reducing atherothrombotic events. For decades, cardiology has been dominated by broad-application drugs like statins, beta-blockers, and antiplatelet agents, where a "one-size-fits-all" approach has been the standard.
If Dalcetrapib is ultimately approved, it would become the first precision medicine of its kind in the field, validating a new development strategy: the co-development of a cardiovascular drug with a companion diagnostic test.[6] This would set a powerful precedent, encouraging the re-examination of other failed cardiovascular trials for hidden pharmacogenomic signals and promoting the integration of genetics into the design of future clinical trials from their inception. The journey of Dalcetrapib, from a symbol of a failed hypothesis to a potential beacon for a new era of personalized medicine, illustrates the dynamic and often unpredictable nature of scientific progress.
Published at: October 11, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.