MedPath

Lapaquistat Advanced Drug Monograph

Published:Sep 22, 2025

Generic Name

Lapaquistat

Drug Type

Small Molecule

Chemical Formula

C31H39ClN2O8

CAS Number

189059-71-0

Lapaquistat (TAK-475): A Comprehensive Pharmacological and Clinical Retrospective of a Squalene Synthase Inhibitor

Executive Summary

Lapaquistat, developed by Takeda Pharmaceutical Company under the code TAK-475, represents a significant chapter in the history of lipid-lowering drug development. It emerged as a first-in-class squalene synthase inhibitor, a novel therapeutic strategy conceived to circumvent the known limitations of statins, particularly statin-associated muscle symptoms. The core of its design was a downstream inhibition of the cholesterol biosynthesis pathway, a mechanistically elegant approach intended to reduce cholesterol production without depleting essential non-sterol isoprenoids. Extensive Phase 2 and 3 clinical trials, involving over 6,000 patients, confirmed that Lapaquistat effectively lowered low-density lipoprotein cholesterol (LDL-C) and other atherogenic markers in a dose-dependent manner. However, this efficacy was overshadowed by a critical safety signal. At the most effective therapeutic dose of 100 mg, Lapaquistat was associated with a significant incidence of elevated liver transaminases, culminating in two cases that met the criteria for Hy's Law—a strong predictor of severe drug-induced liver injury. This dose-limiting hepatotoxicity, coupled with the insufficient efficacy of the safer 50 mg dose, rendered the drug commercially non-viable, leading to the termination of its development program in 2008. Despite this clinical failure, the story of Lapaquistat did not end. Post-hoc research has unveiled remarkable potential for its repurposing. Its precise on-target mechanism is now being explored as a novel host-directed therapy for the parasitic disease cryptosporidiosis and as a potential treatment for the rare genetic inflammatory disorder, Mevalonate Kinase Deficiency. Lapaquistat thus serves as a compelling case study in rational drug design, the unforeseen complexities of metabolic intervention, and the enduring scientific value of even discontinued compounds.

I. Molecular Profile and Physicochemical Properties of Lapaquistat

A precise understanding of the chemical nature of Lapaquistat and its orally administered prodrug, Lapaquistat acetate, is fundamental to interpreting its pharmacological activity and clinical trial data. The development program utilized the prodrug to optimize pharmacokinetic properties, which was then rapidly converted to the active pharmacological agent in vivo.

Identification and Nomenclature

The compound is known by several names across research, clinical, and database contexts, with a critical distinction between the prodrug and its active metabolite.

Lapaquistat (Active Metabolite):

  • Generic Name: Lapaquistat [1]
  • Synonyms: T-91485, T 91485 [1]
  • DrugBank ID: DB16215 [2]
  • CAS Number: 189059-71-0 [1]
  • ChEMBL ID: CHEMBL341976 [2]

Lapaquistat Acetate (Prodrug):

  • Synonyms: TAK-475, TAK 475 [6]
  • DrugBank ID: DB05317 [9]
  • CAS Number: 189060-13-7 [3]
  • Developmental Sponsor: Takeda Pharmaceutical Company Limited [5]

Chemical Structure and Stereochemistry

Lapaquistat is a complex small molecule belonging to the benzoxazepine class of compounds.[13] Its structure contains specific stereocenters that are critical for its biological activity.

Lapaquistat (Active Metabolite):

  • Molecular Formula: C31​H39​ClN2​O8​ [2]
  • Molecular Weight: Approximately 603.1 g/mol [2]
  • IUPAC Name: 2-acetyl]piperidin-4-yl]acetic acid [2]
  • Structural Identifiers:
  • InChIKey: HDGUKVZPMPJBFK-LEAFIULHSA-N [2]
  • SMILES: CC(C)(CN1C2=C(C=C(C=C2)Cl)[C@H](O[C@@H](C1=O)CC(=O)N3CCC(CC3)CC(=O)O)C4=C(C(=CC=C4)OC)OC)CO [2]

Lapaquistat Acetate (Prodrug):

  • Molecular Formula: C33​H41​ClN2​O9​ [8]
  • Molecular Weight: Approximately 645.14 g/mol [8]
  • Structural Difference: The prodrug form is an ester, featuring an acetate group attached to the hydroxyl of the 3-hydroxy-2,2-dimethylpropyl side chain. This modification enhances its oral bioavailability, and the ester is readily cleaved in vivo to release the active Lapaquistat molecule.[8]

Key Physicochemical Properties

The physical and chemical properties of Lapaquistat influence its behavior in biological systems and its handling for research purposes.

  • Solubility: Lapaquistat is reported to be soluble in dimethyl sulfoxide (DMSO) at a concentration of 25 mg/mL (approximately 41.45 mM).[4] The prodrug, Lapaquistat acetate, exhibits higher solubility in DMSO at 46 mg/mL (71.30 mM).[15]
  • Physical Appearance: The compound is described as a white to off-white solid.[18]
  • Predicted Properties: Computational models predict a low water solubility of 0.0205 mg/mL and a logP value of approximately 3.2, indicating a lipophilic nature that facilitates passage across cell membranes to reach its intracellular target enzyme.[19]

The table below consolidates the primary identifiers and properties of both the active metabolite and the prodrug, providing a clear reference for the two key chemical entities discussed throughout this report.

AttributeLapaquistat (Active Metabolite)Lapaquistat Acetate (Prodrug)
Generic NameLapaquistatLapaquistat Acetate
Common SynonymT-91485TAK-475
CAS Number189059-71-0189060-13-7
DrugBank IDDB16215DB05317
Molecular FormulaC31​H39​ClN2​O8​C33​H41​ClN2​O9​
Molecular Weight603.11 g/mol645.14 g/mol

II. Pharmacology: A Novel Mechanism in Cholesterol Synthesis Inhibition

The development of Lapaquistat was predicated on a sophisticated pharmacological hypothesis aimed at creating a new class of lipid-lowering agents with a superior safety profile compared to the established HMG-CoA reductase inhibitors (statins). This involved a deliberate targeting of an enzyme positioned strategically within the cholesterol biosynthesis pathway.

The Cholesterol Biosynthesis Pathway

Cholesterol synthesis is a complex, multi-step process known as the mevalonate pathway. The enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase catalyzes an early, rate-limiting step—the conversion of HMG-CoA to mevalonate.[7] This is the target of statins. Following the formation of mevalonate, the pathway proceeds through several intermediates, including a series of isoprenoids such as farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP). At the point of FPP, the pathway branches. One branch continues toward the synthesis of non-sterol molecules essential for various cellular functions, such as protein prenylation (using FPP and GGPP), mitochondrial respiration (ubiquinone/Coenzyme Q10), and protein glycosylation (dolichol).[21] The other branch is committed exclusively to the synthesis of cholesterol.[21]

The Squalene Synthase Target

Lapaquistat's molecular target is the enzyme squalene synthase (SQS), also known as farnesyl-diphosphate farnesyltransferase (FDFT1).[1] SQS catalyzes the head-to-head condensation of two molecules of FPP to form squalene.[2] The profound significance of this enzymatic step is that it represents the

first committed step in the pathway that is solely dedicated to the production of cholesterol.[2] By inhibiting SQS, Lapaquistat blocks the flow of precursors into the sterol-specific branch of the pathway, thereby reducing the de novo synthesis of cholesterol. This reduction in intracellular cholesterol is expected to upregulate hepatic LDL receptors, leading to increased clearance of LDL-C from the circulation, a mechanism shared with statins.[21]

Comparative Pharmacology: Lapaquistat vs. HMG-CoA Reductase Inhibitors (Statins)

The rationale for developing Lapaquistat becomes clear when its mechanism is contrasted with that of statins. This comparison highlights a deliberate attempt at rational drug design to engineer a safer therapeutic agent.

  • Statins (Upstream Inhibition): By inhibiting HMG-CoA reductase, statins act far upstream in the pathway. This blockade reduces the synthesis of mevalonate and, consequently, all downstream products. This includes not only cholesterol but also the entire family of essential non-sterol isoprenoids (FPP, GGPP, etc.).[20]
  • Lapaquistat (Downstream Inhibition): By inhibiting SQS, Lapaquistat acts downstream of the branch point for isoprenoid synthesis. This was hypothesized to achieve the desired reduction in cholesterol without depleting the cellular pool of FPP and GGPP.[5] In fact, due to the enzymatic block and subsequent feedback mechanisms that upregulate HMG-CoA reductase activity, this mechanism was expected to cause an accumulation, or "backup," of upstream intermediates like FPP.[13]

The central premise of the Lapaquistat program was that it could uncouple the therapeutic effect of cholesterol reduction from the potential side effects associated with isoprenoid depletion. Statin-associated muscle symptoms (SAMS), a significant cause of statin discontinuation, have been mechanistically linked in some preclinical models to the depletion of these isoprenoids, which are vital for skeletal muscle cell health and mitochondrial function.[7] Therefore, targeting SQS was a direct strategy to create a lipid-lowering drug that would not cause myotoxicity.[7]

However, this elegant hypothesis carried an inherent and ultimately critical risk. The fundamental consequence of inhibiting any enzyme is the accumulation of its direct substrate. By blocking SQS, Lapaquistat was designed to cause a buildup of FPP. While this was intended to preserve isoprenoid-dependent functions, it also presented the cell with a significant metabolic challenge: how to handle this excess FPP. The cell may shunt these excess precursors into alternative metabolic pathways. It has been proposed that this accumulation could lead to the formation of potentially toxic metabolites, such as farnesol-derived dicarboxylic acids, which could contribute to cellular stress and organ damage.[24] Thus, the very strategy designed to mitigate one form of toxicity—myotoxicity—inadvertently created the conditions for another, unforeseen toxicity in the liver, the primary site of the drug's action. The attempt to solve the problem of isoprenoid depletion created the new problem of substrate accumulation.

The following table provides a direct mechanistic comparison, clarifying the foundational differences in pharmacological strategy between Lapaquistat and statins.

FeatureHMG-CoA Reductase Inhibitors (Statins)Squalene Synthase Inhibitors (Lapaquistat)
Target EnzymeHMG-CoA ReductaseSqualene Synthase (FDFT1)
Location in PathwayEarly, rate-limiting step (Upstream)First committed step to cholesterol (Downstream)
Effect on CholesterolDecreased synthesisDecreased synthesis
Effect on Isoprenoids (FPP, GGPP)Decreased synthesisSpared or increased due to substrate backup
Hypothesized Primary Safety AdvantageNone (known risk of myotoxicity)Avoidance of myotoxicity by sparing isoprenoids

III. Clinical Pharmacokinetics and Metabolism

The pharmacokinetic profile of Lapaquistat acetate (TAK-475) was a key element of its clinical development, designed to ensure adequate delivery of the active compound to its hepatic target. The data from preclinical and clinical studies reveal a well-defined process of absorption, metabolism, distribution, and excretion.

The Prodrug Strategy: Absorption and Metabolism

Lapaquistat was administered orally as its acetate prodrug, TAK-475, a common strategy to improve the absorption of a parent molecule.[23] Following oral administration, TAK-475 is rapidly absorbed but undergoes extensive first-pass metabolism. Studies in animal models demonstrated that the majority of the TAK-475 prodrug is hydrolyzed to its active metabolite, Lapaquistat (referred to in pharmacokinetic studies as M-I or T-91485), during the absorption process within the intestinal wall.[25]

This efficient conversion has a profound impact on the drug's bioavailability. The systemic bioavailability of the intact parent prodrug (TAK-475) was found to be quite low in animal models, measured at 3.5% in rats and 8.2% in dogs.[25] However, the primary circulating radioactive component in plasma was the active metabolite, M-I (Lapaquistat).[25] This indicates that while the parent drug itself does not reach high systemic concentrations, its active form does. These pharmacokinetic characteristics observed in animals were later confirmed to be consistent in human clinical studies.[25]

Distribution and Hepatic Concentration

A critical feature of Lapaquistat's pharmacokinetics is its distribution to the liver. Despite the low systemic bioavailability of the parent prodrug, concentrations of the active metabolite M-I were found to be much higher in the liver—the main site of cholesterol biosynthesis and the drug's intended organ of action—than in the plasma.[26] This preferential accumulation in the target tissue is a highly desirable pharmacokinetic property, as it maximizes on-target activity while potentially minimizing systemic exposure and off-target effects. While the specific human transporters responsible for this hepatic uptake are not fully detailed, studies indicate that organic cation transporter 2 (OCT2) is not involved.[27]

This profile highlights a crucial distinction between systemic bioavailability and target site engagement. A low bioavailability figure for a parent prodrug can be misleading if not considered in the context of its conversion to an active metabolite and that metabolite's distribution. In the case of Lapaquistat, the low bioavailability of TAK-475 was not a marker of failure but rather an indicator of an efficient first-pass conversion process that successfully delivered the active pharmacological agent to the systemic circulation and, more importantly, concentrated it at the site of action. This confirms that the observed clinical efficacy and toxicity were direct consequences of on-target pharmacology in the liver, driven by adequate drug exposure, rather than being artifacts of poor or erratic pharmacokinetics.

Elimination Profile

The primary route of elimination for Lapaquistat and its metabolites is through fecal excretion.[23] In rats, the absorbed radioactivity was shown to be mainly excreted into the bile, predominantly as the active metabolite M-I. This biliary-excreted M-I was also found to be partially subject to enterohepatic circulation, where it could be reabsorbed from the intestine and returned to the liver, potentially prolonging its duration of action.[25] The overall pharmacokinetic profile was deemed compatible with a once-daily dosing regimen in clinical trials.[23]

IV. The Lapaquistat Clinical Development Program: A Retrospective

The clinical development of Lapaquistat acetate by Takeda was a large-scale, global effort, reflecting the high expectations for a novel lipid-lowering agent with a potentially superior safety profile. The program was designed to rigorously evaluate the drug's efficacy and safety both as a monotherapy and in combination with existing standards of care.

Program Scope

The development program was extensive, encompassing a total of 39 clinical studies that spanned from Phase 1 to advanced Phase 3 trials.[28] Data analysis from the core Phase 2 and 3 studies involved a pooled population of 6,151 patients.[24] These trials were conducted between 2002 and 2007 at numerous sites across the United States, Canada, Europe, Russia, South America, and South Africa, underscoring the global scale of the investigation.[28]

Study Designs and Patient Populations

The trials were designed according to rigorous standards, typically as randomized, double-blind, parallel-group studies with either placebo or active controls. Study durations varied from 6 weeks to a long-term safety study of 96 weeks.[28] The program explored Lapaquistat's utility in several key clinical contexts:

  • Monotherapy Trials: Several studies evaluated Lapaquistat acetate as a standalone therapy. These trials compared various doses of Lapaquistat (typically 25 mg, 50 mg, and 100 mg daily) against placebo and, in some cases, against active comparators like atorvastatin and simvastatin.[23] Key examples include trials NCT00532558 and NCT00865228.[1]
  • Combination Therapy Trials: A significant portion of the program focused on evaluating Lapaquistat as an add-on therapy for patients inadequately controlled on other lipid-lowering drugs. This strategy is common for new agents entering a well-established market. Lapaquistat was studied in combination with:
  • Statins: Including simvastatin (e.g., NCT00286481, NCT00256178) and atorvastatin.[23]
  • Fibrates: Specifically with fenofibrate (e.g., NCT00813527).[6]
  • Cholesterol Absorption Inhibitors: Co-administration with ezetimibe was also investigated.[28]
  • Patient Populations: The trials enrolled a broad range of patients with dyslipidemia, including those with primary hypercholesterolemia and mixed dyslipidemia.[28] The program also included studies in high-risk, difficult-to-treat populations, such as patients with heterozygous and homozygous familial hypercholesterolemia (HoFH), a severe genetic disorder characterized by extremely high LDL-C levels.[1]

The table below summarizes some of the key clinical trials that formed the backbone of the Lapaquistat development program, illustrating the breadth of the clinical investigation.

Trial IdentifierPhasePrimary IndicationStudy DesignKey Investigated Doses
NCT005325583HypercholesterolemiaMonotherapy vs. Placebo50 mg
NCT005323113HypercholesterolemiaCombination with Statins vs. Placebo + Statins50 mg
NCT00286481Not specifiedHypercholesterolemiaCombination with Simvastatin (Factorial)50 mg, 100 mg
NCT002630813Homozygous Familial Hypercholesterolemia (HoFH)Combination with existing lipid-lowering therapyNot specified
NCT00813527Not specifiedHypercholesterolemiaCombination with Fenofibrate100 mg
NCT008652282HypercholesterolemiaMonotherapy (Dosing Regimen) vs. Placebo100 mg (AM vs. PM vs. 50 mg BID)

V. Analysis of Clinical Efficacy

The extensive clinical trial program for Lapaquistat generated a robust dataset on its ability to modify lipid profiles and other biomarkers associated with cardiovascular risk. The results consistently demonstrated that Lapaquistat was a biologically active and effective agent, though its potency relative to the standard of care was a key consideration.

Primary Efficacy: LDL-Cholesterol Reduction

The primary goal of Lapaquistat was to lower LDL-C, and it succeeded in this measure with statistical significance and a clear dose-response relationship.[5]

  • Monotherapy Performance: When used as a standalone treatment, Lapaquistat 100 mg daily reduced LDL-C levels by a mean of 21.6% to 26.3% compared to placebo over 8 to 12 weeks.[23] The 50 mg daily dose resulted in a more modest LDL-C reduction of approximately 18%, while the 25 mg dose produced a 15.8% reduction.[21]
  • Combination Therapy Performance: As an add-on to stable statin therapy, Lapaquistat provided significant additional LDL-C lowering. The 100 mg dose reduced LDL-C by a further 18.0% to 19.1% beyond the effect of the statin alone.[21] This demonstrated a complementary mechanism of action and positioned Lapaquistat as a potential option for patients unable to reach their LDL-C goals on statins alone.
  • Potency Compared to Statins: While effective, Lapaquistat's potency for LDL-C reduction was not as great as that of commonly used statins. In a head-to-head monotherapy comparison, Lapaquistat 100 mg achieved a 26.3% reduction in LDL-C, whereas atorvastatin 10 mg, a standard starting dose, achieved a more robust 36.4% reduction in the same patient population.[23]

Effects on Other Lipid and Inflammatory Markers

Beyond LDL-C, Lapaquistat demonstrated favorable effects on a range of other cardiovascular risk markers.

  • Other Lipoproteins: The drug produced statistically significant reductions in other atherogenic lipid parameters, including total cholesterol (TC), non-HDL-cholesterol, apolipoprotein B (ApoB), very-low-density lipoprotein cholesterol (VLDL-C), and triglycerides (TGs).[20]
  • High-Density Lipoprotein Cholesterol (HDL-C): An interesting and potentially beneficial finding was Lapaquistat's effect on HDL-C. In contrast to atorvastatin, which had a minimal effect, Lapaquistat at doses of 50 mg and 100 mg produced statistically significant increases in HDL-C, with mean increases of 7.7% and 6.3%, respectively, in one study.[23]
  • Inflammatory Markers: Lapaquistat was also shown to reduce levels of high-sensitivity C-reactive protein (hsCRP), a key marker of systemic inflammation and an independent predictor of cardiovascular events. This reduction was observed to be dose-dependent, suggesting a potential anti-inflammatory effect beyond its lipid-lowering properties.[21]

The table below summarizes the pooled efficacy data from key trials, providing a quantitative overview of Lapaquistat's impact on lipid and inflammatory parameters as both monotherapy and in combination with statins.

ParameterLapaquistat 50 mg Monotherapy (% Change)Lapaquistat 100 mg Monotherapy (% Change)Atorvastatin 10 mg Monotherapy (% Change)Lapaquistat 100 mg + Statin (% Add-on Change)
LDL-C-18.4-26.3-36.4-19.1
Total Cholesterol-11.8-16.6-24.4-13.2
HDL-C+7.7+6.3+4.8+1.6
Apolipoprotein B-15.3-19.6-29.6-15.9
Triglycerides-17.2-14.1-19.3-5.8
hsCRPSignificant Reduction (Dose-dependent)Significant Reduction (Dose-dependent)Not specifiedSignificant Reduction

Data synthesized from sources.[20]

VI. The Critical Safety Signal: Unraveling the Hepatotoxicity and Discontinuation

While the efficacy data for Lapaquistat were promising, the ultimate fate of the drug was determined by its safety profile. A critical, dose-dependent safety signal emerged from the extensive clinical program, leading to a re-evaluation of the drug's risk-benefit profile and the eventual termination of its development.

Overall Adverse Event Profile

Across the pooled studies, the overall incidence of adverse events was slightly higher for patients receiving Lapaquistat compared to placebo. However, the types of individual events reported were generally similar between groups and consistent with those seen in this patient population. Common adverse events included headache, nasopharyngitis, myalgia, and back pain.[23] The rate of withdrawal from studies due to adverse events was higher in the Lapaquistat groups (5.7%) compared to the placebo/control groups (3.1%).[35]

A central component of the drug's development hypothesis was improved muscle safety relative to statins. However, the clinical trial data did not bear this out. There was no discernible advantage for Lapaquistat regarding muscle-related side effects; the overall incidence of such events was low and comparable across all treatment arms, including statin monotherapy.[21] The initial pharmacological hypothesis of a myopathy-sparing mechanism was not validated in the large-scale clinical setting.

The Hepatotoxicity Signal

The decisive factor that halted the Lapaquistat program was the emergence of a clear signal of potential hepatic toxicity, which was strongly associated with the 100 mg dose.[2]

  • Elevated Transaminases: The clinical data revealed a dose-dependent increase in the liver enzyme alanine aminotransferase (ALT). The incidence of clinically significant ALT elevations—defined as a value greater than or equal to three times the upper limit of normal (≥3x ULN) on two or more consecutive visits—was markedly higher with Lapaquistat 100 mg. In the pooled efficacy studies, this occurred in 2.0% of patients on Lapaquistat 100 mg versus only 0.3% on placebo. In a long-term safety study, the rate was 2.7% for Lapaquistat 100 mg compared to 0.7% for low-dose atorvastatin.[24]
  • The Hy's Law Cases: The most alarming finding was that two patients receiving Lapaquistat 100 mg met the criteria for Hy's Law. This is a regulatory benchmark used to identify drugs with a high potential to cause severe, life-threatening drug-induced liver injury (DILI). Hy's Law is fulfilled when a drug causes hepatocellular injury (indicated by elevated ALT) combined with evidence of impaired liver function (indicated by elevated total bilirubin), in the absence of cholestasis (elevated alkaline phosphatase).[20] Even a small number of such cases in a pre-market database is considered a very serious safety concern, as it predicts a higher rate of acute liver failure if the drug were to be used in a larger population. This finding represented an unacceptable risk and was the definitive signal that made further development of the 100 mg dose untenable.

The Decision to Terminate Development

Faced with this clear evidence, Takeda Pharmaceutical Company announced the discontinuation of the entire Lapaquistat development program on March 28, 2008.[5] The official statement noted that the decision was based on a thorough review of all available clinical data and the judgment that the compound's overall profile was not superior to existing marketed drugs from both an efficacy and safety standpoint.[12]

This decision was not based on safety alone but was the result of a complex risk-benefit and commercial assessment. The drug was effectively trapped in a commercially unviable position. The 100 mg dose, which offered clinically meaningful efficacy, carried an unacceptable risk of severe hepatotoxicity. The 50 mg dose, which appeared to have a much cleaner safety profile without the same risk of liver enzyme elevation, offered an LDL-C reduction (~18%) that was insufficient to compete effectively in a market dominated by potent and increasingly generic statins capable of achieving >35% LDL-C reduction at standard doses.[23] Neither development path presented a viable option. Pursuing the 100 mg dose would have faced immense regulatory hurdles and post-market risk, while pursuing the 50 mg dose would have meant launching a new, branded drug with inferior efficacy to the established standard of care. This placed Lapaquistat in a pharmaco-economic "valley of death," where the therapeutic window did not align with a commercially viable market position, making the decision to terminate the program a logical, albeit disappointing, conclusion.

The table below highlights the key safety findings that underpinned this decision.

Safety OutcomePlacebo / ControlLapaquistat 50 mgLapaquistat 100 mg
Withdrawal due to Adverse Event3.1%Not specified5.7%
Myalgia/Muscle-related Adverse EventsNo significant difference across groupsNo significant difference across groupsNo significant difference across groups
ALT ≥3x ULN (consecutive visits)~0.3%No similar risk noted2.0% - 2.7%

Footnote: Two cases of Hy's Law (concurrent ALT and bilirubin elevation) were reported in patients receiving Lapaquistat 100 mg. Data synthesized from sources.[21]

VII. Post-Hoc Insights and Future Perspectives for a Discontinued Compound

Although Lapaquistat failed to reach the market for its intended indication, its well-characterized mechanism and extensive dataset have provided invaluable lessons for drug development and have opened surprising new avenues for therapeutic repurposing. The legacy of Lapaquistat is evolving from a story of clinical failure to one of scientific opportunity.

Lessons in Drug Development

The Lapaquistat program serves as a powerful case study in modern pharmacology. It demonstrates that even a highly rational, hypothesis-driven approach to drug design can yield unexpected and unfavorable outcomes. The effort to engineer away the myotoxicity of statins by targeting a downstream enzyme was mechanistically sound, but it failed to account for the full physiological consequences of perturbing a central metabolic pathway—namely, the toxic potential of substrate accumulation.[24] This experience underscores the immense complexity of cellular biochemistry and the limitations of predicting clinical outcomes from preclinical hypotheses alone. Furthermore, the program's termination highlights the challenging commercial landscape for new drugs in established therapeutic areas. A new agent must demonstrate not just efficacy, but a clear and compelling superiority in either efficacy or safety to justify its place against entrenched, cost-effective standards of care.

Emerging Indication: Repurposing for Cryptosporidiosis

Perhaps the most remarkable development in the post-discontinuation story of Lapaquistat is its potential as a treatment for cryptosporidiosis, a severe diarrheal disease caused by the parasite Cryptosporidium.[1] This new application stems from the discovery of a unique host-pathogen interaction. The parasite, an obligate intracellular organism, has evolved to depend on the host cell's metabolic machinery for its survival. Specifically, research has shown that

Cryptosporidium survival hinges on the host cell's production of squalene, the very metabolite synthesized by the enzyme SQS.[1] The accumulation of squalene within the host intestinal epithelial cell creates a reducing environment, making more reduced glutathione available for the parasite to import.

Cryptosporidium has lost the ability to synthesize its own glutathione and is therefore completely dependent on this host-derived supply.[1]

Lapaquistat intervenes directly at this point of dependency. By inhibiting the host's SQS enzyme, Lapaquistat blocks the production of squalene. This prevents the parasite from creating the favorable reducing environment it needs, effectively starving it of essential glutathione and blocking its growth. This effect has been demonstrated both in vitro and in vivo.[1]

This application represents a paradigm shift in anti-infective strategy. Instead of targeting the pathogen directly—a strategy that often leads to the development of drug resistance—Lapaquistat is used as a host-directed therapy. It modulates a host pathway that the pathogen has co-opted, making the host cell an inhospitable environment. The very "on-target" effect that led to chronic hepatotoxicity in hypercholesterolemia patients becomes a potent therapeutic mechanism for an acute infectious disease. The risk-benefit calculation is entirely different; the potential for liver damage from a short course of treatment for a life-threatening infection is far more acceptable than for the chronic management of high cholesterol.

Potential Application in Rare Inflammatory Diseases

Another promising avenue for repurposing involves the rare genetic autoinflammatory disorder Mevalonate Kinase Deficiency (MKD).[36] MKD is caused by mutations in an enzyme that functions early in the mevalonate pathway. This genetic block leads to a deficiency of downstream isoprenoids like FPP and GGPP, which have important anti-inflammatory roles. The resulting isoprenoid deficiency is believed to drive the recurrent fevers and systemic inflammation characteristic of the disease.[13]

The therapeutic hypothesis for Lapaquistat in MKD is a fascinating inversion of its toxic mechanism. By inhibiting SQS, which is located downstream of the isoprenoids, Lapaquistat would cause a metabolic "backup," leading to the accumulation of FPP.[36] In a patient with MKD, this induced accumulation could potentially replenish the depleted pool of FPP, restoring its availability for essential anti-inflammatory signaling pathways and thereby ameliorating the disease symptoms. In vitro studies using cellular models of MKD have already shown that Lapaquistat can exert anti-inflammatory effects, lending support to this hypothesis.[36]

This potential application highlights the duality of a pharmacological effect. The accumulation of FPP, which was a metabolic liability in the context of hypercholesterolemia, could become a therapeutic asset in the context of MKD. It is a powerful illustration of how a deep mechanistic understanding of a drug's action, even one that caused its failure, can unlock its potential for precision medicine in a completely different disease defined by an opposing metabolic imbalance.

VIII. Conclusion and Final Assessment

Lapaquistat (TAK-475) stands as a pivotal and highly informative case study in the pursuit of novel therapeutics for dyslipidemia. Conceived from a sophisticated, rational design intended to create a "safer statin," its journey from a promising pharmacological concept to a discontinued late-stage asset offers critical lessons for the pharmaceutical sciences. The drug's mechanism as a squalene synthase inhibitor successfully achieved its primary goal of lowering LDL-cholesterol and other atherogenic lipoproteins. However, the foundational hypothesis—that by targeting a downstream step in cholesterol synthesis it would avoid the side effects of upstream inhibition—was only partially correct. While it did not appear to have a differential risk of myotoxicity compared to statins, its on-target action led to a different and ultimately unacceptable safety liability: dose-dependent hepatotoxicity.

The termination of the Lapaquistat program was a pragmatic decision driven by the convergence of this safety signal with an insufficient efficacy profile at safer doses, rendering it unable to compete in the demanding cardiovascular market. The experience illustrates with stark clarity that altering central metabolic pathways can produce unforeseen consequences and that the bar for new entrants in a field with effective, established therapies is exceptionally high.

Yet, the conclusion of its development for hypercholesterolemia was not the end of Lapaquistat's scientific relevance. The precise and well-understood mechanism of squalene synthase inhibition has found new life in post-hoc research. The potential for Lapaquistat to be repurposed as a host-directed therapy for cryptosporidiosis and as a metabolic modulator for Mevalonate Kinase Deficiency showcases a modern principle of drug development: a failed drug is not necessarily a failed molecule. The very mechanism that proved detrimental in one chronic, mass-market disease context may be perfectly suited for an acute or rare disease with a different underlying pathophysiology and risk-benefit calculus. Ultimately, the story of Lapaquistat is a testament to the complex, unpredictable nature of clinical translation and a powerful example of how scientific inquiry can continue to extract value and find new therapeutic potential long after a drug's initial development has ceased.

Works cited

  1. Lapaquistat Acetate - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 22, 2025, https://synapse.patsnap.com/drug/4147d7ea57744f90bb5cbfb4432cf962
  2. Lapaquistat | C31H39ClN2O8 | CID 9960389 - PubChem, accessed September 22, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/9960389
  3. Lapaquistat | CAS#189059-71-0 | squalene synthase inhibitor - MedKoo Biosciences, accessed September 22, 2025, https://www.medkoo.com/products/25249
  4. Lapaquistat | TargetMol, accessed September 22, 2025, https://www.targetmol.com/compound/lapaquistat
  5. Lapaquistat - Wikipedia, accessed September 22, 2025, https://en.wikipedia.org/wiki/Lapaquistat
  6. Study Details | NCT00813527 | Effect of Lapaquistat Acetate Combined With Fenofibrate on Blood Cholesterol Levels | ClinicalTrials.gov, accessed September 22, 2025, https://www.clinicaltrials.gov/study/NCT00813527
  7. Effect of Lapaquistat Acetate Combined With Fenofibrate on Blood Cholesterol Levels, accessed September 22, 2025, https://clinicaltrials.gov/study/NCT00813527
  8. Lapaquistat acetate | CAS#189060-13-7 | Squalene Synthase Inhibitor | MedKoo, accessed September 22, 2025, https://www.medkoo.com/products/10341
  9. Fenofibrate Completed Phase 2 Trials for Hyperlipidemias Treatment | DrugBank Online, accessed September 22, 2025, https://go.drugbank.com/drugs/DB01039/clinical_trials?conditions=DBCOND0031916&phase=2&purpose=treatment&status=completed
  10. LAPAQUISTAT ACETATE - precisionFDA, accessed September 22, 2025, https://precision.fda.gov/ginas/app/ui/substances/6673f348-c136-474d-9b48-158ee77cec9b
  11. LAPAQUISTAT CAS#: 189060-13-7 - ChemicalBook, accessed September 22, 2025, https://m.chemicalbook.com/ProductChemicalPropertiesCB01509119_EN.htm
  12. Discontinuation of Development of TAK-475, a Compound for Treatment of Hypercholesterolemia | Fierce Biotech, accessed September 22, 2025, https://www.fiercebiotech.com/biotech/discontinuation-of-development-of-tak-475-a-compound-for-treatment-of-hypercholesterolemia
  13. Anti-inflammatory and cytoprotective effects of a squalene synthase inhibitor, TAK-475 active metabolite-I, in immune cells simulating mevalonate kinase deficiency (MKD)-like condition - PubMed Central, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5005254/
  14. Lapaquistat from Aladdin Scientific | Biocompare.com, accessed September 22, 2025, https://www.biocompare.com/11119-Chemicals-and-Reagents/21049830-Lapaquistat/
  15. Lapaquistat acetate | CAS NO.:189060-13-7 | GlpBio, accessed September 22, 2025, https://www.glpbio.com/lapaquistat-acetate.html
  16. KEGG DRUG: Lapaquistat acetate, accessed September 22, 2025, https://www.genome.jp/dbget-bin/www_bget?dr:D06609
  17. Lapaquistat | CAS NO.:189059-71-0 - GlpBio, accessed September 22, 2025, https://www.glpbio.com/gc38457.html
  18. Lapaquistat (T-91485) | Cholesterol Biosynthesis Inhibitor | MedChemExpress, accessed September 22, 2025, https://www.medchemexpress.com/lapaquistat.html
  19. Lapaquistat: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 22, 2025, https://go.drugbank.com/drugs/DB16215
  20. Squalene Synthase Inhibitor Lapaquistat Acetate | Circulation - AHA Journals, accessed September 22, 2025, https://www.ahajournals.org/doi/10.1161/CIRCULATIONAHA.111.028571
  21. Squalene Synthase Inhibitor Lapaquistat Acetate: Could Anything Be Better Than Statins?, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3807098/
  22. Lapaquistat acetate, a squalene synthase inhibitor, changes macrophage/lipid-rich coronary plaques of hypercholesterolaemic rabbits into fibrous lesions - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2451058/
  23. Lowering LDL Cholesterol -- Beyond Statins - Medscape Education, accessed September 22, 2025, https://www.medscape.org/viewarticle/551353
  24. Lapaquistat Acetate Development of a Squalene Synthase Inhibitor for the Treatment of Hypercholesterolemia | Request PDF - ResearchGate, accessed September 22, 2025, https://www.researchgate.net/publication/51075939_Lapaquistat_Acetate_Development_of_a_Squalene_Synthase_Inhibitor_for_the_Treatment_of_Hypercholesterolemia
  25. Pharmacokinetics of TAK-475, a Squalene Synthase Inhibitor, in Rats and Dogs - PubMed, accessed September 22, 2025, https://pubmed.ncbi.nlm.nih.gov/26839207/
  26. Pharmacokinetics of TAK-475, a Squalene Synthase Inhibitor, in Rats and Dogs, accessed September 22, 2025, https://www.thieme-connect.de/products/ejournals/abstract/10.1055/s-0035-1569407?device=desktop&innerWidth=412&offsetWidth=412&lang=en
  27. Pharmacokinetics properties of geraniin, corilagin, ellagic acid, and lapaquistat, accessed September 22, 2025, https://www.researchgate.net/figure/Pharmacokinetics-properties-of-geraniin-corilagin-ellagic-acid-and-lapaquistat_tbl3_340853019
  28. Lapaquistat Acetate | Circulation - American Heart Association Journals, accessed September 22, 2025, https://www.ahajournals.org/doi/10.1161/circulationaha.110.975284
  29. Lapaquistat Acetate | Circulation - AHA Journals, accessed September 22, 2025, https://www.ahajournals.org/doi/abs/10.1161/circulationaha.110.975284
  30. Lapaquistat acetate: development of a squalene synthase inhibitor for the treatment of hypercholesterolemia - PubMed, accessed September 22, 2025, https://pubmed.ncbi.nlm.nih.gov/21518985/
  31. Study Details | NCT00532558 | Efficacy of Lapaquistat Acetate on Blood Cholesterol Levels in Treating Subjects With Hypercholesterolemia | ClinicalTrials.gov, accessed September 22, 2025, https://clinicaltrials.gov/study/NCT00532558
  32. Study Details | NCT00286481 | Efficacy of Lapaquistat Acetate Alone or Combined With Simvastatin in Subjects With Hypercholesterolemia | ClinicalTrials.gov, accessed September 22, 2025, https://www.clinicaltrials.gov/ct2/keydates/NCT00286481
  33. Study Details | NCT00286481 | Efficacy of Lapaquistat Acetate Alone or Combined With Simvastatin in Subjects With Hypercholesterolemia, accessed September 22, 2025, https://www.clinicaltrials.gov/study/NCT00286481
  34. Lapaquistat Terminated Phase 3 Trials for High Cholesterol Treatment | DrugBank Online, accessed September 22, 2025, https://go.drugbank.com/drugs/DB16215/clinical_trials?conditions=DBCOND0037835&phase=3&purpose=treatment&status=terminated
  35. Lapaquistat Acetate - AHA Journals, accessed September 22, 2025, https://www.ahajournals.org/doi/pdf/10.1161/circulationaha.110.975284
  36. Mevalonate Kinase Deficiency and Squalene Synthase Inhibitor (TAK-475): The Balance to Extinguish the Inflammation - MDPI, accessed September 22, 2025, https://www.mdpi.com/2218-273X/11/10/1438

Published at: September 22, 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.