Small Molecule
C19H27NO3
105816-04-4
Type 2 Diabetes Mellitus
Nateglinide is an orally administered, short-acting insulin secretagogue belonging to the meglitinide (or glinide) class of antidiabetic agents. Chemically, it is a D-phenylalanine derivative, distinguishing it structurally from the sulfonylurea class of drugs.[1] Its primary therapeutic application is as an adjunct to diet and exercise for the management of type 2 diabetes mellitus, specifically targeting postprandial hyperglycemia.[3] The pharmacological action of nateglinide is characterized by a unique "fast on-fast off" kinetic profile at its molecular target, the pancreatic β-cell ATP-sensitive potassium channel.[5] This property enables it to mimic the physiological first-phase insulin secretion that occurs in response to a meal, leading to rapid but transient insulin release that effectively controls post-meal glucose excursions.
The clinical use of nateglinide is governed by its rapid and short-lived action. A strict, meal-dependent dosing regimen is essential for both efficacy and safety, encapsulated by the critical patient counseling point: "skip a meal, skip a dose".[3] This approach mitigates the principal adverse effect of hypoglycemia. Pharmacokinetically, nateglinide is characterized by rapid absorption, a short half-life of approximately 1.5 hours, and extensive hepatic metabolism. Its clearance is predominantly mediated by the cytochrome P450 isozyme CYP2C9, a fact that introduces significant considerations for pharmacogenomics and drug-drug interactions, particularly in individuals who are genetically poor metabolizers of the enzyme.[7] While approved by major regulatory bodies, including the U.S. Food and Drug Administration (FDA), its marketing authorization in the European Union was withdrawn in 2022 for commercial reasons, reflecting a shift in the therapeutic landscape toward newer antidiabetic agents with more convenient dosing and broader metabolic benefits.[9]
Nateglinide represents a targeted approach to the management of type 2 diabetes mellitus, a complex metabolic disorder characterized by insulin resistance and progressive β-cell dysfunction. It is classified as an oral antihyperglycemic agent within the meglitinide (glinide) class, a group of non-sulfonylurea insulin secretagogues known for their rapid onset and short duration of action.[1]
Structurally, nateglinide is an N-acylated derivative of the amino acid D-phenylalanine, specifically (2R)-3-phenyl-2-({[trans-4-(1-methylethyl)cyclohexyl]carbonyl}amino)-3-phenylpropanoic acid.[2] This chemical identity distinguishes it fundamentally from the sulfonylurea and biguanide classes of antidiabetic drugs. This structural uniqueness is directly responsible for its distinct pharmacokinetic and pharmacodynamic properties, which define its therapeutic niche.
The primary role of nateglinide in diabetes management is that of a prandial glucose regulator. Its mechanism is finely tuned to restore the deficient first-phase insulin response, a key pathophysiological defect in type 2 diabetes that leads to exaggerated postprandial glucose excursions.[1] By stimulating a rapid, short-lived pulse of insulin secretion timed with meal ingestion, nateglinide addresses this specific defect. This targeted action contrasts sharply with longer-acting secretagogues like sulfonylureas, which provide a more sustained, meal-independent insulin release, or agents like metformin, which primarily target basal hepatic glucose production. This positions nateglinide not as a foundational, broad-spectrum glucose-lowering agent, but as a specialist drug for patients whose primary glycemic challenge is the control of blood sugar after meals.
A precise understanding of nateglinide's chemical and physical properties is fundamental to its formulation, pharmacology, and biopharmaceutical behavior.
Nateglinide is a white to off-white crystalline powder.[8] Its solubility profile reflects a lipophilic character, which is consistent with its oral absorption and extensive hepatic metabolism. It is practically insoluble in water but exhibits good solubility in organic solvents: freely soluble in methanol, ethanol, and chloroform; soluble in ether; and sparingly soluble in acetonitrile and octanol.[8] The melting point is reported to be in the range of 137–141 °C.[20]
For unambiguous identification across various scientific and regulatory databases, a comprehensive list of identifiers is provided in Table 1.
Table 1: Comprehensive Drug Identifiers for Nateglinide
Identifier Type | Identifier Value | Source/Database |
---|---|---|
DrugBank ID | DB00731 | DrugBank 1 |
CAS Number | 105816-04-4 | ChemIDplus, DrugBank 1 |
UNII | 41X3PWK4O2 | FDA Global Substance Registration System (GSRS) 1 |
PubChem CID | 5311309 | PubChem 2 |
ChEBI ID | CHEBI:31897 | Chemical Entities of Biological Interest 1 |
KEGG ID | D01111 | Kyoto Encyclopedia of Genes and Genomes 1 |
European Community (EC) Number | 642-283-9 | European Chemicals Agency (ECHA) 1 |
ATC Code | A10BX03 | WHO Collaborating Centre for Drug Statistics Methodology 20 |
RxCUI | 274332 | NLM RxNorm Terminology 1 |
The therapeutic effect of nateglinide is derived from a precise and well-characterized pharmacological mechanism that modulates pancreatic insulin secretion in a manner that closely mimics the body's natural response to a meal.
Nateglinide's primary pharmacological action is the stimulation of insulin secretion from pancreatic β-cells, an effect that is contingent upon the presence of functioning β-cells.[3] The molecular target for this action is the ATP-sensitive potassium (
KATP) channel on the β-cell membrane.[8] Nateglinide binds to the sulfonylurea receptor 1 (SUR1) subunit of this channel, a protein also identified as the ATP-binding cassette sub-family C member 8 (ABCC8).[5]
The binding of nateglinide to the SUR1 subunit closes the KATP channel, inhibiting the outward flow of potassium ions. This leads to a depolarization of the β-cell membrane. The change in membrane potential triggers the opening of voltage-gated calcium channels, resulting in an influx of extracellular calcium (Ca2+). The subsequent rise in intracellular calcium concentration is the final signal that initiates the exocytosis of pre-formed, insulin-containing secretory granules, releasing insulin into the bloodstream.[8]
While the general mechanism of closing KATP channels is shared with sulfonylureas, nateglinide's pharmacodynamic profile is distinguished by several unique features that define its clinical utility.
The most critical of these is its binding kinetics, often described as a "fast on-fast off" effect.[5] Nateglinide associates with the SUR1 receptor rapidly but with a relatively low affinity, and, most importantly, it dissociates from the receptor extremely quickly, on the order of seconds.[5] This kinetic profile is the molecular basis for its rapid onset of action (stimulating insulin secretion within 20 minutes) and very short duration of action (insulin levels return to baseline within approximately 4 hours).[11]
This rapid, transient action allows nateglinide to preferentially stimulate and restore the physiological first-phase of insulin secretion, the initial, sharp peak of insulin release that occurs within minutes of nutrient ingestion.[5] In type 2 diabetes, this first phase is characteristically blunted or absent, leading to poor initial control of meal-derived glucose. By restoring this early insulin peak, nateglinide effectively attenuates the subsequent postprandial glucose spike.[5]
Furthermore, the insulinotropic effect of nateglinide is glucose-dependent. Its ability to stimulate insulin release diminishes as blood glucose levels fall.[8] Unlike sulfonylureas, which can continue to promote insulin secretion even at hypoglycemic levels, nateglinide has little to no effect on insulin release in the absence of glucose.[24] This glucose-sensing mechanism is a key safety feature, reducing the risk of interprandial (between-meal) and fasting hypoglycemia.
When compared to other insulin secretagogues, nateglinide's profile is distinct. Its action is more rapid and of shorter duration than that of both traditional sulfonylureas (e.g., glyburide) and the fellow meglitinide repaglinide.[5] This leads to superior control of postprandial glucose excursions and a lower risk of delayed hyperinsulinemia, which can cause hypoglycemia hours after a meal.[5] However, this specialization in prandial control comes with a trade-off. Clinical studies have demonstrated that nateglinide is generally less effective than metformin or glyburide at lowering integrated measures of long-term glycemic control, such as glycosylated hemoglobin (
HbA1c) and fasting plasma glucose (FPG).[13] The entire clinical utility and safety profile of nateglinide can thus be understood as a direct consequence of its unique binding kinetics at the SUR1 receptor. The rapid "on" rate necessitates its administration just before a meal to align insulin release with glucose absorption. The rapid "off" rate and short half-life ensure its effect is transient, covering only the mealtime glucose load and minimizing the risk of delayed hypoglycemia, which in turn dictates the "skip a meal, skip a dose" rule. Finally, its glucose-dependent action provides an intrinsic safety mechanism against severe hypoglycemia, distinguishing it from older secretagogues.
The pharmacokinetic profile of nateglinide is characterized by rapid absorption, extensive protein binding, efficient hepatic metabolism, and swift elimination, all of which are consistent with its role as a short-acting prandial glucose regulator.
Following oral administration, nateglinide is rapidly and completely absorbed from the gastrointestinal tract, with an estimated absolute bioavailability of approximately 73%.[8] Peak plasma concentrations (
Cmax) are typically achieved within one hour after dosing, allowing for a rapid onset of its pharmacodynamic effect.[8]
The timing of administration relative to food intake is a critical factor. When nateglinide is taken with or after a meal, the rate of its absorption is delayed, as evidenced by a decrease in Cmax and a prolonged time to peak concentration (Tmax); however, the overall extent of absorption (AUC) is not significantly affected.[8] Studies have shown that administration just prior to food consumption (e.g., 10 to 30 minutes before) results in optimal pharmacodynamic effects, with a more rapid insulin response and better control of the postprandial glucose peak.[27]
Nateglinide has a relatively small steady-state volume of distribution (Vd), estimated to be approximately 10 liters in healthy subjects.[8] It is extensively bound (98%) to plasma proteins, primarily serum albumin and, to a lesser extent,
α1-acid glycoprotein.[8] This high degree of protein binding suggests a low potential for displacement interactions with other highly protein-bound drugs, as confirmed in in vitro studies.[13]
Nateglinide undergoes extensive metabolism in the liver via the cytochrome P450 (CYP) mixed-function oxidase system prior to elimination.[1] The primary metabolic pathways involve hydroxylation of the isopropyl group or the cyclohexyl ring, followed by glucuronide conjugation.[8]
The key enzymes responsible for its metabolism are well-defined: nateglinide is predominantly metabolized by CYP2C9 (approximately 70%) and to a lesser extent by CYP3A4 (approximately 30%).[7] This heavy reliance on CYP2C9 is a critical aspect of its clinical pharmacology, forming the basis for significant drug interactions and pharmacogenomic variability.
The major metabolites produced are less potent hypoglycemic agents than the parent compound. However, a minor metabolite, the isoprene metabolite, possesses a potency similar to that of nateglinide itself.[1]
Nateglinide and its metabolites are rapidly and completely eliminated from the body. Following an oral dose, approximately 83% is recovered in the urine and 10% in the feces.[1] The majority of the renally excreted portion consists of metabolites; only about 16% of the dose is excreted in the urine as unchanged parent drug.[8]
The elimination half-life (t1/2) of nateglinide is very short, averaging approximately 1.5 hours.[8] This rapid elimination prevents drug accumulation with multiple daily doses and reinforces its short duration of action, ensuring that its glucose-lowering effect is confined to the postprandial period.
Table 2: Key Pharmacokinetic Parameters of Nateglinide
Parameter | Value | Reference(s) |
---|---|---|
Absolute Bioavailability (F) | ~73% | 8 |
Time to Peak (Tmax) | <1 hour | 8 |
Plasma Protein Binding | 98% | 8 |
Volume of Distribution (Vd) | ~10 L | 8 |
Elimination Half-life (t1/2) | ~1.5 hours | 8 |
Primary Metabolic Pathway | Hepatic (CYP2C9: 70%, CYP3A4: 30%) | 7 |
Primary Excretion Route | Renal (83% of dose, primarily as metabolites) | 1 |
The clinical utility of nateglinide is precisely defined by its pharmacological profile, positioning it as a targeted therapy for postprandial hyperglycemia in type 2 diabetes.
Nateglinide is indicated as an adjunct to diet and exercise to improve glycemic control in adults with type 2 diabetes mellitus.[3] It can be used as monotherapy or in combination with other oral antidiabetic agents, most notably metformin.[11]
The drug's mechanism of action dictates its limitations. Nateglinide is explicitly contraindicated and not for use in patients with type 1 diabetes or for the treatment of diabetic ketoacidosis (DKA).[3] These conditions are characterized by an absolute or near-absolute deficiency of insulin, and since nateglinide requires functioning pancreatic β-cells to exert its effect, it would be ineffective. Insulin therapy is required for these conditions.
In clinical trials involving patients with type 2 diabetes previously treated with diet alone (treatment-naïve), nateglinide monotherapy demonstrated statistically significant reductions in mean HbA1c and fasting plasma glucose (FPG) levels compared to placebo.[8] However, its potency in this regard is modest compared to other established agents. Head-to-head comparisons have shown nateglinide to be less effective than either metformin or the second-generation sulfonylurea glyburide at lowering these overall measures of glycemic control.[13]
Nateglinide's role is often more pronounced in combination therapy. When added to metformin in patients inadequately controlled on metformin alone, nateglinide provides a significant additive effect, leading to greater reductions in both HbA1c and FPG than could be achieved with either agent as monotherapy.[5] This synergy arises from their complementary mechanisms of action: metformin primarily reduces hepatic glucose production and improves insulin sensitivity, while nateglinide directly addresses prandial insulin secretion.
Conversely, a clinical trial examining the addition of nateglinide to a regimen of a patient inadequately controlled on glyburide found no additional clinical benefit.[13] This finding suggests a mechanistic redundancy; since both drugs are insulin secretagogues acting on the
KATP channel, adding nateglinide to a maximally effective dose of a sulfonylurea does not further enhance insulin release.
This body of clinical evidence paints a clear picture of nateglinide's role. It is not a first-line, "generalist" agent for achieving powerful, broad-spectrum glucose lowering. Its relative weakness in reducing FPG and overall HbA1c compared to workhorse drugs like metformin makes it less suitable as a foundational monotherapy. Instead, its strength as a "specialist" in targeting postprandial hyperglycemia defines its place in therapy. It is a logical add-on for a patient who is on metformin but continues to experience significant glucose spikes after meals. This targeted application leverages its unique pharmacodynamic profile without relying on it for 24-hour glycemic control.
Effective and safe use of nateglinide is highly dependent on strict adherence to its specific dosing and administration schedule, which requires thorough patient education.
Nateglinide is available for oral administration as tablets in two strengths: 60 mg and 120 mg.[18]
The timing of nateglinide administration is critical to its mechanism and safety.
Comprehensive patient counseling is paramount for the safe and effective use of nateglinide. Key counseling points include:
While generally well-tolerated when used appropriately, nateglinide has a distinct safety profile dominated by the risk of hypoglycemia.
The principal and most serious risk associated with nateglinide therapy is hypoglycemia (low blood sugar).[3]
Nateglinide is strictly contraindicated in the following patient populations:
The potential for drug interactions with nateglinide is significant, arising from both pharmacokinetic (metabolism-based) and pharmacodynamic (effect-based) mechanisms. The central role of CYP2C9 in its clearance makes it particularly vulnerable to interactions with inhibitors and inducers of this enzyme.
Table 3: Clinically Significant Drug Interactions with Nateglinide
Interacting Drug/Class | Mechanism of Interaction | Potential Clinical Effect | Clinical Recommendation |
---|---|---|---|
Fluconazole, Amiodarone | Strong CYP2C9 Inhibition | Increased nateglinide levels; high risk of severe hypoglycemia | Monitor closely; consider dose reduction 7 |
Rifampin, Carbamazepine, Barbiturates | CYP2C9 Induction | Decreased nateglinide levels; loss of glycemic control | Monitor for efficacy; may require dose increase 31 |
Efavirenz, Ivosidenib | CYP2C9 Inhibition/Induction | Significant alteration of nateglinide levels | Avoid or use alternate drug 31 |
Other Antidiabetics (e.g., Metformin, SGLT2i) | Pharmacodynamic Synergism | Additive hypoglycemic effect | Monitor closely; may require dose reduction of one agent 31 |
NSAIDs, Salicylates, MAOIs | Potentiation of Hypoglycemia | Increased risk of hypoglycemia | Monitor closely 7 |
Thiazides, Corticosteroids | Pharmacodynamic Antagonism | Decreased hypoglycemic effect; hyperglycemia | Monitor for loss of glycemic control 11 |
Non-selective Beta-Blockers | Masks adrenergic symptoms of hypoglycemia | Unawareness of developing hypoglycemia | Counsel patient; increase frequency of glucose monitoring 11 |
Alcohol | Potentiation of Hypoglycemia | Severe, potentially prolonged hypoglycemia | Avoid or limit consumption; contraindicated with excessive use 31 |
Grapefruit Juice | CYP Enzyme Inhibition | Increased nateglinide levels; increased risk of adverse effects | Avoid consumption 33 |
The use of nateglinide requires careful consideration in specific patient populations and in those with certain genetic predispositions.
The metabolism of nateglinide presents a clinically significant example of a gene-drug interaction.
This pharmacogenomic consideration elevates the discussion of nateglinide's safety beyond general warnings. It identifies a specific, genetically defined patient subpopulation at high risk for its primary dose-limiting toxicity. This is not merely a theoretical concern but a practical clinical issue that underscores the importance of personalized medicine. The heavy reliance of nateglinide on a single, highly polymorphic enzyme for its clearance makes it a candidate for preemptive pharmacogenetic testing to enhance prescribing safety.
Nateglinide was developed by the Japanese company Ajinomoto and marketed globally by Novartis under various brand names, most notably Starlix.[14] Other brand names include Starsis, Trazec, and Fastic.[1]
The U.S. Food and Drug Administration (FDA) granted approval for nateglinide (Starlix) in December 2000 for the management of type 2 diabetes.[13] While the original brand name product, Starlix, has since been discontinued, generic versions of nateglinide remain available on the U.S. market.[1]
The European Medicines Agency (EMA) granted marketing authorization for nateglinide under the brand names Starlix and Trazec on April 3, 2001.[1] The indication in Europe was specifically for combination therapy with metformin in patients inadequately controlled on metformin alone.[9] However, on April 29, 2022, the marketing authorization for Starlix was withdrawn from the European Union at the request of the manufacturer, Novartis, which cited
commercial reasons for the decision.[9] The authorization for Trazec had previously lapsed under the "sunset clause" due to it never being marketed in Europe.[41]
While specific approval dates from Australia's Therapeutic Goods Administration (TGA) are not detailed in the available materials, the drug's approval and availability in that market are confirmed by the assignment of an AU TGA Pregnancy Category C.[37]
The commercial withdrawal of nateglinide from the competitive European market, despite its established efficacy in a specific clinical niche, is telling. This decision was likely a consequence of its clinical profile when compared to the newer classes of antidiabetic agents that have emerged since its launch. Nateglinide's demanding thrice-daily dosing regimen, which is strictly tied to meals, represents a significant burden on patient adherence. Furthermore, its primary adverse effects of hypoglycemia and weight gain are precisely the liabilities that newer drug classes—such as DPP-4 inhibitors, SGLT-2 inhibitors, and GLP-1 receptor agonists—have sought to minimize or even reverse. Most critically, nateglinide has no proven benefit on macrovascular outcomes, whereas several newer agents have demonstrated cardiovascular risk reduction, a paramount goal in modern diabetes care.[18] In a crowded therapeutic landscape, a drug with a higher treatment burden, a less favorable side-effect profile, and no demonstrated cardiovascular benefit becomes commercially less viable, even if it remains mechanistically effective. Its clinical profile, therefore, appears to have directly influenced its market longevity.
Nateglinide is a pharmacologically elegant drug, representing a second generation of insulin secretagogues designed with a specific therapeutic goal: to address postprandial hyperglycemia by restoring the deficient first-phase insulin secretion characteristic of type 2 diabetes. Its unique "fast on-fast off" binding kinetics at the pancreatic KATP channel and its glucose-dependent action are significant refinements over older, long-acting sulfonylureas, offering a more physiological approach to mealtime glucose control with a theoretically lower risk of interprandial hypoglycemia.
However, the very properties that define its specialized role also impose its clinical limitations. The short half-life and rapid action necessitate a demanding and relatively inflexible thrice-daily, meal-dependent dosing regimen, which can be a barrier to long-term patient adherence. Despite its more refined mechanism, its use still carries an inherent and clinically significant risk of hypoglycemia, as well as a tendency for weight gain.
From a contemporary clinical perspective, while nateglinide remains an effective tool for a specific patient profile—namely, an individual on metformin with isolated postprandial hyperglycemia—its place in the broader diabetes treatment algorithm has been largely superseded. The advent of newer drug classes, including DPP-4 inhibitors, SGLT-2 inhibitors, and GLP-1 receptor agonists, has shifted the paradigm of diabetes management. These modern agents offer the convenience of once-daily or even weekly dosing, are associated with a much lower risk of hypoglycemia, have neutral or beneficial effects on body weight, and, in the case of SGLT-2 inhibitors and GLP-1 receptor agonists, have demonstrated robust cardiovascular and renal benefits.
Ultimately, the story of nateglinide serves as an important case study in drug development and the evolution of therapeutic standards. It highlights how a clinically effective medication can become less relevant as the treatment landscape evolves to prioritize not only glycemic efficacy but also patient convenience, safety from hypoglycemia, and the reduction of long-term cardiovascular complications. The most enduring clinical lesson from nateglinide may be the critical importance of understanding its CYP2C9-mediated metabolism and the associated pharmacogenomic variability, which stands as a clear and actionable principle for ensuring patient safety in an era of personalized medicine.
Published at: September 26, 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.