MedPath

Lansoprazole Advanced Drug Monograph

Published:May 29, 2025

Generic Name

Lansoprazole

Brand Names

Prevacid, Prevpac

Drug Type

Small Molecule

Chemical Formula

C16H14F3N3O2S

CAS Number

103577-45-3

Associated Conditions

Duodenal Ulcer, Erosive Esophagitis, Helicobacter Pylori Infection, Symptomatic Gastroesophageal Reflux Disease (sGERD), Ulcer Healing, Zollinger-Ellison Syndrome, Active Duodenal ulcer, Develop NSAID-induced gastric ulcers, Hypersecretory conditions, Maintenance of healing Erosive esophagitis

Lansoprazole: A Comprehensive Pharmacological and Clinical Review

1. Introduction and Overview

Lansoprazole is a widely utilized pharmaceutical agent belonging to the class of proton pump inhibitors (PPIs). It is primarily prescribed for the management of conditions related to excessive gastric acid secretion.

  • Generic Name: Lansoprazole [1]
  • Brand Names: Lansoprazole is marketed under various brand names globally, with Prevacid® being one of the most recognized. Other common names include Prevacid 24HR (an over-the-counter formulation), Prevacid SoluTab (an orally disintegrating tablet), and Prevacid OTC.[1] During its development, it was also known by the code AG 1749.[1] The availability of multiple brand names, including over-the-counter (OTC) options, reflects its extensive clinical use and established profile for certain indications. However, this also necessitates careful patient education to prevent inadvertent duplication of therapy or inappropriate self-medication if a patient is also prescribed a PPI.
  • DrugBank ID: DB00448 [1]
  • CAS Number: 103577-45-3 [1]
  • Chemical Type: Lansoprazole is a small molecule drug [User Query]. Structurally, it is classified as a substituted benzimidazole.[1] It exists as a racemic 1:1 mixture of its two enantiomers, dexlansoprazole (the R-enantiomer) and levolansoprazole (the S-enantiomer).[1] The benzimidazole core is a common structural feature among many PPIs, which may imply shared mechanistic properties and, theoretically, a potential for cross-reactivity in hypersensitivity reactions, although specific data on lansoprazole cross-reactivity were not detailed in the provided materials. The existence of dexlansoprazole as a separately marketed, enantiomerically pure drug suggests potential pharmacokinetic or pharmacodynamic distinctions from the racemic mixture.
  • Brief Summary of Use and Therapeutic Class: As a proton pump inhibitor, lansoprazole effectively reduces the production of stomach acid.[1] Its primary therapeutic applications include the treatment of peptic ulcer disease (both duodenal and gastric ulcers), gastroesophageal reflux disease (GERD), and Zollinger-Ellison syndrome. It is also a key component in multi-drug regimens for the eradication of Helicobacter pylori infection when implicated in ulcerative disease.[1]
  • Historical Background and Development: Lansoprazole was originally synthesized by Takeda Pharmaceuticals and was assigned the developmental code AG 1749.[1] It was patented in 1984 and subsequently launched for medical use in 1991 in Europe and 1992 in Japan.[1] In the United States, lansoprazole received approval from the Food and Drug Administration (FDA) in 1995, becoming the second PPI available in the US market.[1] The patent protection for the lansoprazole molecule expired in November 2009, paving the way for the introduction of generic formulations under various brand names.[1] Since 2009, lower-strength formulations of lansoprazole have been available over-the-counter in the U.S. for the treatment of frequent heartburn, such as Prevacid 24HR and Lansoprazole 24HR.[1] The extensive period of market availability has allowed for the accumulation of substantial clinical experience and post-marketing surveillance data, contributing to a well-characterized efficacy and safety profile. The transition to OTC status for specific, less severe indications further attests to its established safety record when used appropriately for self-management.

2. Pharmacology

2.1. Mechanism of Action

Lansoprazole functions as a prodrug, meaning it is administered in an inactive form and requires conversion to its active moiety within the body.[7] This activation occurs specifically within the highly acidic environment of the secretory canaliculi of gastric parietal cells.[7] Once activated, lansoprazole covalently binds to and irreversibly inhibits the hydrogen/potassium adenosine triphosphatase (H+/K+-ATPase) enzyme system, commonly known as the proton pump.[1] This enzyme is located on the secretory surface of parietal cells and represents the final step in the pathway of gastric acid secretion, responsible for pumping hydrogen ions (H+) into the gastric lumen in exchange for potassium ions (K+).[7]

By forming a covalent disulfide bond with cysteine residues on the H+/K+-ATPase, particularly on the alpha-subunit, lansoprazole effectively blocks the pump's function.[5] This inhibition leads to a profound and sustained reduction in both basal and stimulated gastric acid secretion, irrespective of the physiological stimulus (e.g., histamine, acetylcholine, gastrin).[11] Because the inhibition is irreversible, the suppression of acid secretion persists until new H+/K+-ATPase enzyme units are synthesized by the parietal cells and integrated into the secretory membrane. This process typically takes 24 to 48 hours, explaining the prolonged duration of action of lansoprazole despite its relatively short plasma half-life.[7] The selective accumulation and activation of lansoprazole in the acidic canaliculi of parietal cells contribute to its targeted action and minimize systemic effects.

2.2. Pharmacodynamics

The pharmacodynamic effects of lansoprazole are characterized by a rapid onset and prolonged duration of gastric acid suppression, leading to an increase in intragastric pH.

  • Onset of Action: Inhibition of gastric acid secretion begins relatively quickly after oral administration. An increase in intragastric pH is typically observed within 1 to 2 hours following a 30 mg dose and within 2 to 3 hours following a 15 mg dose.[12] Some sources indicate an onset of gastric acid suppression within 1 to 3 hours generally.[13]
  • Duration of Effect: The acid-suppressing effects of lansoprazole extend for up to a couple of days, a consequence of the irreversible inhibition of the proton pump.[1] Specifically, acid secretion remains suppressed for 24 to 48 hours, as new proton pump synthesis is required to restore acid production.[7] Clinical studies have demonstrated that after 14 days of daily administration of lansoprazole 30 mg, intragastric pH, basal acid output, and stimulated acid output return to baseline levels within 2 to 4 days following discontinuation of the drug. Importantly, this return to baseline occurs without evidence of rebound acid hypersecretion.[14]
  • Quantitative Effects on Gastric pH and Acid Output: Lansoprazole administration leads to a significant dose-dependent decrease in basal acid output and a corresponding significant increase in mean intragastric pH.[12] It also markedly increases the percentage of time during which the intragastric pH remains above critical thresholds, such as pH 3.0 and pH 4.0.[12] Maintaining an intragastric pH above 4.0 is generally considered necessary for the healing of acid-related mucosal lesions and reflects a 99% reduction in gastric acid.[12] In a pharmacodynamic crossover study, lansoprazole 30 mg once daily resulted in a mean 24-hour intragastric pH of 4.53 (compared to a baseline of 2.1). With this dose, the gastric pH was maintained above 3.0 for 75% of the 24-hour period and above 4.0 for 63% of the time.[15] Even a 15 mg once-daily dose significantly increased the mean 24-hour pH to 3.97, with pH >3.0 for 64% of the time and >4.0 for 48% of the time.[15] After multiple daily dosing, the increase in gastric pH is observed within the first hour post-dosing with 30 mg of lansoprazole and within 1 to 2 hours post-dosing with 15 mg.[12] Comparative studies indicate that lansoprazole 30 mg administered nasogastrically achieved significantly higher mean 24-hour intragastric pH values compared to pantoprazole 40 mg IV on both day 1 (pH 3.05 vs. 2.76) and day 5 (pH 3.65 vs. 3.45) of treatment.[16] A meta-analysis assessing the relative potencies of various PPIs based on their effect on mean 24-hour gastric pH determined lansoprazole to have a relative potency of 0.90 when omeprazole was set as the reference at 1.00.[17] This suggests a comparable, though slightly less potent on a mg-per-mg basis, acid-suppressing effect compared to omeprazole in terms of overall pH control.

2.3. Pharmacokinetics

The pharmacokinetic profile of lansoprazole is characterized by rapid absorption, high protein binding, extensive hepatic metabolism primarily via CYP2C19 and CYP3A4, and a relatively short elimination half-life that belies its prolonged pharmacodynamic effect.

  • Absorption: Lansoprazole is rapidly absorbed following oral administration.[13] The bioavailability of oral lansoprazole is generally high, reported as 80% or more.[1] However, the absorption of lansoprazole is significantly affected by food. Administration 30 minutes after a meal can decrease its maximum plasma concentration (Cmax) and the area under the plasma concentration-time curve (AUC) by 50-70%.[13] This substantial food effect underscores the clinical recommendation to administer lansoprazole before meals to ensure optimal absorption and efficacy.[12] The time to reach peak plasma concentration (Tmax) is approximately 1.7 hours.[13]
  • Distribution: Lansoprazole is highly bound to plasma proteins, with a protein binding extent of 97%.[1] The volume of distribution (Vd) has been reported as 0.61 to 0.9 L/kg in children and 15.7 ± 1.9 L in adults.[13]
  • Metabolism: Lansoprazole undergoes extensive metabolism, primarily in the liver.[1] The major metabolic pathways involve the cytochrome P450 (CYP) enzyme system. CYP2C19 is the principal enzyme responsible for the formation of 5'-hydroxy lansoprazole, accounting for over 80% of its metabolism, while CYP3A4 mediates the formation of lansoprazole sulfone.[1] These metabolites are essentially devoid of antisecretory activity.[18] Lansoprazole exhibits linear pharmacokinetics, meaning its plasma concentrations increase proportionally with the dose.[20] The extensive hepatic metabolism, particularly through CYP2C19 which is known for its genetic polymorphisms, is a key factor contributing to inter-individual variability in drug exposure and response.
  • Excretion: The metabolites of lansoprazole are excreted through both renal and fecal/biliary routes. Approximately one-third (33%) of the administered dose is excreted in the urine, with 14-25% as metabolites and less than 1% as unchanged drug.[1] The remaining two-thirds (67%) are eliminated via biliary excretion into the feces.[13] The plasma elimination half-life of lansoprazole is relatively short, typically ranging from 1.0 to 1.5 hours in adults [1], with some reports citing 1.5 ± 1 hour [13] or a mean of 1.11 hours.[14] This short half-life is not directly proportional to the duration of its acid-suppressing effect, which is much longer due to the irreversible nature of its binding to the proton pump.[1] The half-life may be prolonged in older adults (1.9 to 2.9 hours) and in patients with hepatic impairment (4 to 7.2 hours).[13]
  • Pharmacokinetics in Pediatric Populations: The pharmacokinetics of lansoprazole have been studied across various pediatric age groups, revealing age-dependent differences.
  • Children (1-11 years): When dosed at 15 mg daily for those ≤30 kg and 30 mg daily for those >30 kg, the mean Cmax and AUC values on Day 5 were comparable between the two dose groups and were not significantly affected by body weight or age within each dosing cohort. The overall pharmacokinetic profile in this age group was similar to that observed in healthy adult subjects.[12] Reported clearance is 0.57 to 0.71 L/hour/kg, and the half-life is 1.2 to 1.5 hours.[13]
  • Adolescents (12-17 years): Following administration of 15 mg or 30 mg daily, mean Cmax and AUC values were not significantly influenced by body weight or age. Nearly dose-proportional increases in Cmax and AUC were observed between the two doses, and the pharmacokinetic profile was similar to that of healthy adults.[12]
  • Neonates (<28 days) and Infants (1-11 months): Neonates exhibited higher systemic exposure (mean weight-based normalized AUC values were 2.04- and 1.88-fold higher at doses of 0.5 mg/kg/day and 1 mg/kg/day, respectively) compared to healthy adults receiving a 30 mg dose. Infants aged ≤10 weeks showed clearance and exposure values similar to those of neonates. In contrast, infants older than 10 weeks who received lansoprazole at 1 mg/kg/day had mean AUC values comparable to adults receiving a 30 mg dose.[24] The elimination half-life in neonates was reported to be 2-3 hours, which is longer than that observed in older children and adults.[24] These findings reflect the immaturity of drug-metabolizing enzyme systems, particularly CYP2C19, in early life, leading to decreased clearance and increased exposure in the youngest pediatric patients.

3. Clinical Efficacy and Therapeutic Uses

3.1. Approved Indications

Lansoprazole is approved for a range of acid-related disorders in both adult and pediatric populations, as specified below. Its efficacy stems from its potent inhibition of gastric acid secretion.

  • Duodenal Ulcer:
  • Short-Term Treatment of Active Duodenal Ulcer (Adults): The standard dose is 15 mg once daily for 4 weeks.[12]
  • Maintenance of Healed Duodenal Ulcers (Adults): For preventing recurrence, the recommended dose is 15 mg once daily.[12]
  • Helicobacter pylori Eradication to Reduce the Risk of Duodenal Ulcer Recurrence (Adults): Lansoprazole is a component of multi-drug regimens aimed at eradicating H. pylori, a primary cause of duodenal ulcers.[1]
  • Triple Therapy: This typically involves lansoprazole 30 mg twice daily, amoxicillin 1 gram twice daily, AND clarithromycin 500 mg twice daily. The duration of triple therapy is either 10 or 14 days.[12]
  • Dual Therapy: An alternative regimen consists of lansoprazole 30 mg three times daily AND amoxicillin 1 gram three times daily, administered for 14 days.[12] This regimen is indicated for patients who are allergic or intolerant to clarithromycin, or in situations where clarithromycin resistance is known or suspected.[19]
  • For patients with a penicillin allergy, alternative H. pylori eradication regimens are necessary. While specific lansoprazole-based penicillin-free regimens are not detailed in the primary FDA labels, general guidelines recommend combinations such as a PPI (like lansoprazole) plus clarithromycin and metronidazole.[25] The Prevpac® co-packaged product, which contains lansoprazole, amoxicillin, and clarithromycin, is contraindicated in patients with known allergies to amoxicillin or clarithromycin.[27]
  • Benign Gastric Ulcer:
  • Short-Term Treatment (Adults): The usual dosage is 30 mg once daily for up to 8 weeks.[12]
  • NSAID-Associated Gastric Ulcer (Adults): Lansoprazole is indicated for both the healing of gastric ulcers that develop in patients taking nonsteroidal anti-inflammatory drugs (NSAIDs) and for reducing the risk of ulcer development in susceptible patients.[1]
  • Healing: 30 mg once daily for 8 weeks. Controlled studies supporting this indication did not extend beyond 8 weeks.[12]
  • Risk Reduction (in patients with a history of a documented gastric ulcer who require continued NSAID use): 15 mg once daily for up to 12 weeks. Controlled studies for this indication did not extend beyond 12 weeks.[12]
  • Gastroesophageal Reflux Disease (GERD): Lansoprazole is widely used for various manifestations of GERD in both adults and children.[1]
  • Adults:
  • Short-Term Treatment of Symptomatic GERD: For relief of heartburn and other GERD-related symptoms, the dose is 15 mg once daily for up to 8 weeks.[12]
  • Short-Term Treatment of Erosive Esophagitis (EE): For healing of esophageal erosions, the dose is 30 mg once daily for up to 8 weeks. If healing is not achieved within this timeframe (in 5-10% of patients), an additional 8-week course may be considered. An additional 8-week course may also be considered for recurrence of erosive esophagitis.[12]
  • Maintenance of Healing of Erosive Esophagitis: To prevent recurrence of erosions, the dose is 15 mg once daily. Controlled studies for this indication extended up to 12 months.[19]
  • Pediatrics:
  • Children 1 to 11 years (for Symptomatic GERD and Short-Term Treatment of Erosive Esophagitis): Dosing is weight-based.[12]
  • Patients weighing ≤30 kg: 15 mg once daily for up to 12 weeks.
  • Patients weighing >30 kg: 30 mg once daily for up to 12 weeks.
  • In some pediatric patients who remained symptomatic after 2 or more weeks of initial therapy, the dose was increased (up to 30 mg twice daily).[12]
  • Children 12 to 17 years (for Short-Term Treatment of Symptomatic GERD):.[12]
  • Non-erosive GERD: 15 mg once daily for up to 8 weeks.
  • Erosive Esophagitis: 30 mg once daily for up to 8 weeks.
  • Pathological Hypersecretory Conditions Including Zollinger-Ellison Syndrome (ZES) (Adults): This group of disorders is characterized by excessive gastric acid production.[1]
  • The recommended adult starting dose is 60 mg once daily. Doses should be individualized based on patient needs and therapy should continue as long as clinically indicated. Dosages up to 90 mg twice daily have been administered. If the total daily dose exceeds 120 mg, it should be administered in divided doses. Some patients with ZES have been treated continuously with lansoprazole for more than four years.[19]
  • Over-the-Counter (OTC) Use (Adults): Nonprescription lansoprazole (15 mg) is indicated for the treatment of frequent heartburn, defined as heartburn that occurs two or more days per week.[10]

The wide array of indications for lansoprazole, spanning acute treatment, maintenance therapy, and risk reduction across different age groups and etiological factors (such as H. pylori infection or NSAID use), underscores its established efficacy and versatility in managing conditions driven by gastric acid. The specific dosing regimens, particularly for H. pylori eradication, are based on combination therapy principles critical for successful outcomes and minimizing resistance. The availability of pediatric dosing information reflects dedicated clinical investigation in these younger populations.

3.2. Comparative Efficacy

The clinical efficacy of lansoprazole has been compared to other PPIs and different drug classes across its various indications. Generally, lansoprazole demonstrates comparable efficacy to other PPIs for many conditions, though some studies suggest nuances in specific outcomes or patient populations.

  • General Comparison with other PPIs: Multiple reviews and guidelines suggest that the clinical differences between various PPIs, including lansoprazole, omeprazole, pantoprazole, and rabeprazole, are often minimal when used at equipotent doses for conditions like GERD and peptic ulcer disease.1 The American Academy of Family Physicians (AAFP) has noted that these PPIs produce comparable rates of healing and remission in erosive esophagitis 32 and that all PPIs are equally effective at equipotent dosages.33 A comprehensive review by Therapeutics Letter concluded there is no convincing randomized controlled trial (RCT) evidence that one PPI is definitively preferable to another for managing GERD or PUD symptoms or for the endoscopic healing of esophagitis, though it highlighted potential biases in many comparative studies.31
  • Gastroesophageal Reflux Disease (GERD) / Erosive Esophagitis (EE):
  • Lansoprazole vs. Omeprazole: For non-erosive reflux disease (NERD), a systematic review found no significant difference in relieving heartburn and regurgitation, although omeprazole appeared more effective in terms of overall gastric acidity control (intragastric pH).[34] Conversely, another meta-analysis suggested that lansoprazole demonstrated superior "quality effectiveness" for GERD symptom relief compared to omeprazole, emphasizing that dose was a critical factor influencing outcomes.[35]
  • Lansoprazole vs. Other PPIs (Esomeprazole, Pantoprazole, Rabeprazole) in EE:
  • In a study involving elderly patients (over 65 years) with EE, pantoprazole 40 mg and rabeprazole 20 mg were found to be significantly more effective than omeprazole 20 mg in healing esophagitis. Lansoprazole 30 mg showed a trend towards better healing rates (90.7%) compared to omeprazole (81.0%), though this difference was not statistically significant (p=0.143). For symptom relief (heartburn, acid regurgitation, epigastric pain), pantoprazole and rabeprazole were more effective than both lansoprazole and omeprazole in this elderly cohort.[36]
  • A network meta-analysis (NMA) comparing FDA-recommended doses for EE indicated that esomeprazole 40 mg demonstrated superiority over lansoprazole 30 mg in mucosal erosion healing at both 4 weeks (Odds Ratio 1.30) and 8 weeks (OR 1.37), as well as in heartburn relief (OR 1.29).[38] Another NMA also concluded that esomeprazole 40 mg daily was the most efficient for healing among various PPIs and H2-receptor antagonists (H2RAs).[39]
  • A direct comparative trial between esomeprazole 40 mg and lansoprazole 30 mg for EE reported significantly higher healing rates with esomeprazole at 8 weeks (92.6% vs. 88.8%) and at 4 weeks. Esomeprazole also led to faster sustained resolution of heartburn, with the difference in healing rates being more pronounced in patients with more severe baseline esophagitis.[40]
  • The American College of Gastroenterology (ACG) 2022 guidelines provide a potency ranking for acid suppression among PPIs (from least to highest): pantoprazole, lansoprazole, omeprazole, esomeprazole, and rabeprazole.[41] This ranking suggests that while all are effective, there are differences in their intrinsic acid-suppressing capabilities that might translate to varying clinical efficacy in certain situations or patient types.
  • Peptic Ulcer (PU):
  • Lansoprazole vs. Omeprazole for H. pylori-associated Duodenal Ulcer (DU): A meta-analysis indicated similar DU healing rates between lansoprazole-containing combination therapies and omeprazole-containing combinations (Relative Risk = 1.04). However, lansoprazole combinations were associated with significantly higher H. pylori eradication rates (RR = 1.09).[42]
  • Lansoprazole vs. Potassium-Competitive Acid Blockers (P-CABs) (e.g., Vonoprazan):
  • A meta-analysis comparing P-CABs to lansoprazole for peptic ulcer healing found that P-CABs demonstrated non-inferiority. Pooled healing rates were 95.3% for P-CABs versus 95.0% for lansoprazole. While treatment-emergent adverse events (TEAEs) were similar, P-CABs were associated with a higher risk of serious adverse events.[43] Another NMA focusing on vonoprazan also concluded its non-inferiority/equivalence to lansoprazole 30 mg for PUD treatment.[47]
  • General PPI Comparison for Gastric Ulcer (GU): A review suggested that newer PPIs, including lansoprazole, pantoprazole, and rabeprazole, demonstrated greater improvement in clinical symptoms compared to omeprazole for gastric ulcers. The pooled RR for PPIs (lansoprazole, omeprazole, pantoprazole) versus ranitidine for GU healing at 4 weeks was 1.33, favoring PPIs.[48]
  • General PPI Comparison for Duodenal Ulcer (DU): An NMA focusing on the initial non-eradication treatment of DU found no significant difference in 4-week ulcer healing rates among various PPIs, including lansoprazole, omeprazole, pantoprazole, rabeprazole, and ilaprazole.[49]

While lansoprazole is a highly effective PPI, the comparative efficacy data suggest that for certain indications like severe erosive esophagitis, or for achieving the most rapid symptom relief, other agents like esomeprazole or the newer P-CABs might offer advantages, albeit sometimes with different safety profiles or cost implications. The choice of PPI should therefore be individualized, considering the specific indication, severity of disease, patient characteristics (including CYP2C19 genotype, as discussed later), potential drug interactions, and cost. The observation that newer PPIs or P-CABs may outperform older ones like omeprazole (and sometimes lansoprazole) in specific metrics highlights the ongoing evolution in acid-suppression therapy.

Table 5: Comparative Efficacy of Lansoprazole vs. Other Acid-Suppressing Agents (Selected Indications)

IndicationComparator Agent(s)Lansoprazole DoseComparator Dose(s)Key Efficacy Outcome (Lansoprazole vs. Comparator)Reference(s)
Erosive Esophagitis (EE) Healing (8 weeks)Omeprazole30 mg/d20 mg/dLansoprazole (90.7%) trended better than Omeprazole (81.0%), p=0.143 (elderly patients)36
EE Healing (8 weeks)Esomeprazole30 mg/d40 mg/dEsomeprazole (92.6%) significantly higher than Lansoprazole (88.8%)40
EE Healing (4 & 8 weeks)Esomeprazole30 mg/d40 mg/dEsomeprazole significantly better healing rates (OR 1.30 at 4w, 1.37 at 8w)38
EE Healing (8 weeks)Pantoprazole30 mg/d40 mg/dPantoprazole (93.5%) vs Lansoprazole (90.7%) - indirect comparison suggests pantoprazole may be slightly higher in this study (elderly)36
EE Healing (8 weeks)Rabeprazole30 mg/d20 mg/dRabeprazole (94.6%) vs Lansoprazole (90.7%) - indirect comparison suggests rabeprazole may be slightly higher in this study (elderly)36
GERD Symptom Relief (Heartburn)OmeprazoleN/AN/ANo significant difference in relieving heartburn/regurgitation (NERD)34
GERD Symptom Relief (Heartburn)Esomeprazole30 mg/d40 mg/dEsomeprazole faster sustained resolution of heartburn40
GERD Symptom Relief (Heartburn)Pantoprazole, Rabeprazole30 mg/d40 mg/d (Panto), 20 mg/d (Rabe)Pantoprazole and Rabeprazole more effective than Lansoprazole for heartburn, regurgitation, epigastric pain (elderly EE patients)36
Duodenal Ulcer Healing (H. pylori associated)Omeprazole (combination therapy)30 mg BID20 mg BID (typical for omeprazole in combos)Similar DU healing rates (RR 1.04)42
H. pylori Eradication Rate (DU)Omeprazole (combination therapy)30 mg BID20 mg BID (typical for omeprazole in combos)Lansoprazole combinations significantly better (RR 1.09)42
Peptic Ulcer Healing (General)P-CABs (e.g., Vonoprazan)30 mg/d (PU treatment)Vonoprazan 20 mg/dP-CABs non-inferior to Lansoprazole (Healing: 95.3% vs 95.0%)43
NSAID-Associated Ulcer Prevention (Recurrence)P-CABs (e.g., Vonoprazan)15 mg/dVonoprazan 10 mg/d or 20 mg/dVonoprazan non-inferior to Lansoprazole46

Note: This table summarizes selected comparative findings. Efficacy can vary based on patient populations, specific study designs, and ulcer/esophagitis grades. "N/A" indicates data was not specific for dose comparison in the cited snippet for that particular outcome.

4. Dosage and Administration

The appropriate dosage and administration of lansoprazole vary depending on the indication, patient age, and specific formulation used. It is crucial to adhere to prescribed dosages and administration instructions to optimize efficacy and safety.

4.1. Formulations

Lansoprazole is available in several oral formulations designed for delayed release to protect the acid-labile drug from degradation in the stomach and ensure delivery to its site of absorption in the small intestine:

  • Delayed-Release Capsules: Available in 15 mg and 30 mg strengths.[1] These capsules are typically opaque, hard gelatin, and may be colored (e.g., pink and green) with brand-specific imprints like "PREVACID 15" or "PREVACID 30".[50]
  • Delayed-Release Orally Disintegrating Tablets (ODT): Marketed under brand names such as Prevacid SoluTab®, these are available in 15 mg and 30 mg strengths.[2] ODTs are designed to dissolve on the tongue before swallowing.
  • Granules for Oral Suspension: Lansoprazole is also available as granules that can be mixed with liquid to form an oral suspension.[7] FDA labels also refer to an oral suspension formulation.[19]
  • Parenteral (Intravenous) Formulation: While some general medical resources mention parenteral use for lansoprazole [1], the provided FDA prescribing information primarily details oral formulations. One study mentions intravenous pantoprazole as a comparator to nasogastrically administered lansoprazole, which might imply that IV lansoprazole is not as commonly used or available in all regions compared to other IV PPIs.[16]

4.2. Adult Dosing

All oral doses of lansoprazole should be taken before eating, ideally 30 to 60 minutes before a meal (best if before breakfast for once-daily dosing) to maximize absorption and efficacy.[12]

  • Active Duodenal Ulcer (Short-Term Treatment): 15 mg once daily for 4 weeks.[12]
  • Maintenance of Healed Duodenal Ulcers: 15 mg once daily.[12]
  • Helicobacter pylori Eradication (to Reduce Duodenal Ulcer Recurrence):
  • Triple Therapy: Lansoprazole 30 mg twice daily (q12h), plus amoxicillin 1 gram twice daily (q12h), plus clarithromycin 500 mg twice daily (q12h), for 10 or 14 days.[12]
  • Dual Therapy (for clarithromycin allergy/intolerance/resistance): Lansoprazole 30 mg three times daily (q8h), plus amoxicillin 1 gram three times daily (q8h), for 14 days.[12]
  • Benign Gastric Ulcer (Short-Term Treatment): 30 mg once daily for up to 8 weeks.[12]
  • NSAID-Associated Gastric Ulcer:
  • Healing: 30 mg once daily for 8 weeks.[12]
  • Risk Reduction: 15 mg once daily for up to 12 weeks.[12]
  • Gastroesophageal Reflux Disease (GERD):
  • Short-Term Treatment of Symptomatic GERD: 15 mg once daily for up to 8 weeks.[12]
  • Short-Term Treatment of Erosive Esophagitis (EE): 30 mg once daily for up to 8 weeks. An additional 8-week course may be considered if healing is not achieved or for recurrence.[12]
  • Maintenance of Healing of Erosive Esophagitis: 15 mg once daily.[19]
  • Pathological Hypersecretory Conditions (including Zollinger-Ellison Syndrome): The recommended adult starting dose is 60 mg once daily. Doses should be individualized based on patient response and continued as clinically indicated. Dosages up to 90 mg twice daily have been administered. Daily doses exceeding 120 mg should be administered in divided doses. Some patients have been treated continuously for more than four years.[19]
  • Heartburn (OTC use): 15 mg once daily for 14 days. This course may be repeated every 4 months if needed.[29]

4.3. Pediatric Dosing

Lansoprazole should also be administered to pediatric patients before eating.[12]

  • Symptomatic GERD and Short-Term Treatment of Erosive Esophagitis (Children 1 to 11 years of age): [12]
  • Patients weighing ≤30 kg: 15 mg once daily for up to 12 weeks.
  • Patients weighing >30 kg: 30 mg once daily for up to 12 weeks.
  • If symptoms persist after 2 or more weeks of initial therapy, the dose may be increased, up to 30 mg twice daily in some patients.[29]
  • Short-Term Treatment of Symptomatic GERD (Adolescents 12 to 17 years of age): [12]
  • Non-erosive GERD: 15 mg once daily for up to 8 weeks.
  • Erosive Esophagitis: 30 mg once daily for up to 8 weeks.
  • Neonates (less than 28 days old) and Infants (1 to 11 months old): While pharmacokinetic studies have been conducted in this age group, showing higher exposure and decreased clearance particularly in neonates and infants ≤10 weeks old [24], the drug manufacturer did not pursue an indication for GERD in infants due to a lack of demonstrated clinical efficacy in studies.[24] Dosing in this very young age group is complex and typically managed by specialists.

4.4. Administration Instructions

  • General: Administer 30 to 60 minutes before a meal; for once-daily dosing, it is best if taken before breakfast. If lansoprazole is administered twice daily, the first dose should be taken before breakfast and the second dose before dinner.[13]
  • Capsules:
  • Swallow capsules whole. Do not split, chew, or crush them.[10]
  • For patients with difficulty swallowing capsules:
  • The capsule may be opened and the intact granules sprinkled on one tablespoon of applesauce, Ensure® pudding, cottage cheese, yogurt, or strained pears. The mixture should then be swallowed immediately without chewing the granules.[10]
  • Alternatively, the capsule may be opened and the contents emptied into approximately 60 mL (2 ounces) of orange juice, apple juice, or tomato juice. Mix briefly and swallow immediately. To ensure complete delivery of the dose, rinse the glass with two or more additional volumes of juice and swallow immediately.[10]
  • Orally Disintegrating Tablets (ODT):
  • Do not break, cut, or chew the ODT.[10]
  • Place the tablet on the tongue and allow it to dissolve (this may take up to one minute) with or without water, until the particles can be swallowed.[10]
  • ODTs may also be administered via an oral syringe: Place a 15 mg tablet in an oral syringe and draw up approximately 4 mL of water, or place a 30 mg tablet in an oral syringe and draw up approximately 10 mL of water. Shake gently to disperse the tablet. Administer the contents within 15 minutes. Refill the syringe with an additional 2 mL of water (for the 15 mg tablet) or 5 mL of water (for the 30 mg tablet), shake gently, and administer any remaining contents.[10]
  • Nasogastric (NG) Tube Administration:
  • Capsule Contents (for NG tubes ≥16 French): To optimize effectiveness, separate administration from enteral nutrition.[13] Open the capsule(s) and disperse the intact granules in 30 to 40 mL of apple juice. Mix briefly. Draw the mixture into an enteral dosing syringe and administer via the NG tube. Rinse the syringe with additional apple juice and administer via the NG tube; repeat the rinse of the syringe and administer to ensure the entire dose is delivered.[13]
  • Orally Disintegrating Tablets (ODT): Some FDA labels state that the ODT product should not be given through enteral administration tubes.[19] However, other sources provide instructions for ODT administration via NG tube (≥8 French) using a method similar to the oral syringe technique (dissolving in water and flushing).[13] This discrepancy highlights a need for clarification based on the specific product formulation and institutional protocols.

4.5. Special Populations

  • Hepatic Impairment:
  • No dosage adjustment is necessary for patients with mild (Child-Pugh Class A) or moderate (Child-Pugh Class B) hepatic impairment.[29]
  • For patients with severe (Child-Pugh Class C) hepatic impairment, a dose reduction should be considered. The FDA prescribing information recommends considering a maximum daily dose of 15 mg for these patients.[12]
  • Renal Impairment: No dosage adjustment is recommended for adult or pediatric patients with renal impairment.[30]

The availability of various formulations, including those suitable for patients with dysphagia or requiring NG tube administration, enhances the clinical utility of lansoprazole. The critical instruction to administer the drug before meals is directly linked to its pharmacokinetic profile, specifically the significant negative impact of food on its bioavailability. Dose adjustments for severe hepatic impairment are essential due to the liver's primary role in lansoprazole metabolism.

Table 1: Lansoprazole Dosage and Administration Summary

IndicationPatient PopulationRecommended DoseFrequencyDuration of TherapySpecial InstructionsReference(s)
Duodenal Ulcer - Active TreatmentAdults15 mgOnce daily4 weeksTake before meals12
Duodenal Ulcer - MaintenanceAdults15 mgOnce dailyAs prescribedTake before meals12
H. pylori Eradication - TripleAdults30 mgTwice daily10 or 14 daysTake before meals12
H. pylori Eradication - DualAdults30 mgThree times daily14 daysTake before meals12
Benign Gastric Ulcer - ActiveAdults30 mgOnce dailyUp to 8 weeksTake before meals12
NSAID-Ulcer - HealingAdults30 mgOnce daily8 weeksTake before meals12
NSAID-Ulcer - Risk ReductionAdults15 mgOnce dailyUp to 12 weeksTake before meals12
Symptomatic GERDAdults15 mgOnce dailyUp to 8 weeksTake before meals12
Erosive Esophagitis - HealingAdults30 mgOnce dailyUp to 8 weeks (may repeat)Take before meals12
Erosive Esophagitis - MaintenanceAdults15 mgOnce dailyUp to 12 months (controlled studies)Take before meals19
Pathological Hypersecretory (ZES)AdultsStarting 60 mgOnce dailyAs clinically indicatedTake before meals; doses >120mg/day divided19
Symptomatic GERD & EE (1-11 yrs)Pediatric (≤30 kg)15 mgOnce dailyUp to 12 weeksTake before meals12
Symptomatic GERD & EE (1-11 yrs)Pediatric (>30 kg)30 mgOnce dailyUp to 12 weeksTake before meals12
Non-erosive GERD (12-17 yrs)Pediatric15 mgOnce dailyUp to 8 weeksTake before meals12
Erosive Esophagitis (12-17 yrs)Pediatric30 mgOnce dailyUp to 8 weeksTake before meals12

EE = Erosive Esophagitis; ZES = Zollinger-Ellison Syndrome. All oral doses should be taken before meals.

5. Safety Profile and Tolerability

Lansoprazole is generally well-tolerated, particularly with short-term use. However, like all medications, it is associated with a range of potential adverse effects, from common and mild to rare but serious. Long-term use carries specific risks that warrant careful consideration and monitoring.

5.1. Common Adverse Reactions

The most frequently reported adverse reactions in short-term, placebo-controlled clinical trials (incidence ≥1%) include [12]:

  • Diarrhea: This is often the most common adverse event, with an incidence of 3.8% for lansoprazole compared to 2.3% for placebo in pooled short-term studies. The incidence was similar between placebo and 15 mg or 30 mg doses of lansoprazole but appeared to be higher with 60 mg doses (7.4%).[12] In a study focusing on NSAID-associated gastric ulcer risk reduction, diarrhea occurred in 5% of lansoprazole-treated patients, compared to 22% with misoprostol and 3% with placebo.[12]
  • Abdominal Pain: Reported in 2.1% of lansoprazole users versus 1.2% in the placebo group.[12]
  • Nausea: Occurred in 1.3% of lansoprazole users compared to 1.2% in the placebo group.[12]
  • Constipation: Reported at an incidence of 1.0% with lansoprazole versus 0.4% with placebo.[12]
  • Headache: While reported at an incidence greater than 1%, headache was more common in the placebo group in some studies.[12]

Other commonly mentioned, generally mild side effects include dry mouth, insomnia, drowsiness, blurred vision, rash, pruritus, dizziness, and flatulence.[1] Some reports also note difficulty achieving or maintaining an erection in men.[10]

5.2. Serious Adverse Reactions

Serious adverse reactions to lansoprazole are rare but can occur. These include:

  • Hypersensitivity Reactions: Anaphylaxis, anaphylactic shock, angioedema, bronchospasm, and urticaria have been reported.[1] Lansoprazole is contraindicated in patients with known severe hypersensitivity to any component of its formulation.[50]
  • Severe Cutaneous Adverse Reactions (SCARs): Rare but potentially life-threatening skin reactions such as Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), bullous eruption, and erythroderma have been associated with lansoprazole use.[1] The FDA label advises discontinuation at the first sign or symptom of SCARs or other signs of hypersensitivity.[28]
  • Acute Interstitial Nephritis (AIN) / Acute Tubulointerstitial Nephritis (ATIN): This has been observed in patients taking PPIs, including lansoprazole. AIN can occur at any point during PPI therapy and is generally considered an idiopathic hypersensitivity reaction. If AIN develops, lansoprazole should be discontinued.[1]
  • Hepatotoxicity: Clinically apparent liver injury due to lansoprazole is rare and typically mild and anicteric. Onset is usually within 2 to 4 weeks of starting therapy, with a hepatocellular or mixed pattern of enzyme elevations. Recovery is generally rapid upon drug withdrawal. Drug Rash with Eosinophilia and Systemic Symptoms (DRESS) syndrome, potentially involving minor liver enzyme elevations, has also been described.[7]
  • Blood Disorders: Rare reports include leukopenia, leukocytosis, pancytopenia, and thrombocytopenia.[1]

Other rarely reported serious events include taste disturbance, peripheral edema, photosensitivity, fever, sweating, depression, arthralgia, and myalgia.[1]

5.3. Long-Term Risks and Precautions

Prolonged use of PPIs, including lansoprazole, has been associated with several potential long-term health risks:

  • Bone Fracture (Osteoporosis-Related): Multiple observational studies suggest that long-term (typically one year or longer) and/or multiple daily dose PPI therapy may be associated with an increased risk of osteoporosis-related fractures of the hip, wrist, or spine.[1] The mechanism is thought to involve impaired calcium absorption due to PPI-induced hypochlorhydria, which can reduce the ionization of dietary calcium salts. This may lead to secondary hyperparathyroidism and increased bone resorption.[51] Another postulated mechanism is the inhibition of osteoclast proton pumps, which are involved in bone remodeling.[53] Patients should use the lowest effective dose for the shortest duration appropriate to their condition.[12] The systemic effect on bone health resulting from a drug primarily targeting gastric acid secretion highlights the complex physiological interconnections within the body.
  • Hypomagnesemia: Symptomatic and asymptomatic hypomagnesemia has been reported rarely in patients treated with PPIs for at least three months, but more commonly after one year of therapy.[1] Serious adverse events of hypomagnesemia can include tetany, arrhythmias, and seizures. Treatment often requires magnesium supplementation and discontinuation of the PPI. For patients expected to be on prolonged PPI treatment or those taking PPIs concomitantly with medications like digoxin or diuretics that can also cause hypomagnesemia, periodic monitoring of serum magnesium levels should be considered, starting prior to initiation of therapy.[12] The proposed mechanism involves impaired gastrointestinal absorption of magnesium, possibly through alteration of intestinal mucosal pH and interference with the transient receptor potential melastatin-6 (TRPM6) magnesium channel.[54] Typically, urinary magnesium excretion is low in affected individuals, indicating renal conservation in response to malabsorption.[54]
  • Clostridioides difficile-Associated Diarrhea (CDAD): PPI therapy, including lansoprazole, may be associated with an increased risk of developing CDAD, particularly in hospitalized patients.[1] CDAD should be considered in any patient on PPIs who presents with persistent diarrhea that does not improve. The lowest effective dose and shortest duration of PPI therapy appropriate for the condition being treated should be used to minimize this risk.[12] The mechanism is believed to be related to the alteration of the gastric pH, which may allow the survival and proliferation of C. difficile spores or vegetative forms that would normally be inactivated by stomach acid. Other potential mechanisms include impairment of leukocyte function or other immune responses by PPIs.[56] This illustrates how local changes in gastric acidity can have downstream consequences on the intestinal microbiome and susceptibility to infection.
  • Cyanocobalamin (Vitamin B12) Deficiency: Daily long-term use of any acid-suppressing medications, including lansoprazole (e.g., for longer than 3 years), may lead to malabsorption of vitamin B12 (cyanocobalamin) due to hypo- or achlorhydria.[7] This diagnosis should be considered if clinical symptoms consistent with vitamin B12 deficiency are observed.[50] Gastric acid and pepsin are essential for cleaving dietary vitamin B12 from food proteins, a necessary step for its subsequent absorption. By reducing gastric acid, PPIs can impair this release, leading to decreased B12 availability.[53]
  • Fundic Gland Polyps: The use of PPIs, particularly long-term, is associated with an increased risk of developing fundic gland polyps.[10] These polyps are generally benign and may regress upon discontinuation of the PPI. While early animal studies with PPIs showed ECL cell proliferation and carcinoid tumors, gastric biopsy specimens from humans treated continuously with lansoprazole for at least one year did not show similar ECL cell effects, though longer-term data are still needed to definitively rule out any increased risk of gastric tumors.[12] The development of these polyps is thought to be driven by the hypergastrinemia that results from chronic acid suppression.[53]
  • Cutaneous and Systemic Lupus Erythematosus: New onset or exacerbation of existing autoimmune diseases, primarily cutaneous lupus erythematosus (CLE) and subacute CLE (SCLE), has been reported with PPI use. If such conditions develop, lansoprazole should be discontinued, and the patient referred to a specialist for evaluation.[7]

5.4. Warnings and Precautions

The FDA-approved prescribing information for lansoprazole includes several important warnings and precautions:

  • Presence of Gastric Malignancy: Symptomatic response to lansoprazole therapy does not preclude the presence of gastric malignancy. Appropriate diagnostic measures should be undertaken if malignancy is suspected, as PPIs can mask symptoms and delay diagnosis.[12]
  • Acute Interstitial Nephritis (AIN) / Acute Tubulointerstitial Nephritis (ATIN): As detailed under serious adverse reactions, AIN has been observed and necessitates discontinuation of the drug.[28]
  • Interactions with Investigations for Neuroendocrine Tumors: Serum chromogranin A (CgA) levels increase secondary to drug-induced hypergastrinemia. Elevated CgA levels may cause false-positive results in diagnostic tests for neuroendocrine tumors. Therefore, lansoprazole treatment should be temporarily stopped for at least 14 days before assessing CgA levels to avoid this interference.[28]

The array of potential long-term risks associated with lansoprazole underscores the importance of judicious prescribing. This includes using the lowest effective dose for the shortest duration necessary, regularly reassessing the indication for continued therapy, and being vigilant for signs and symptoms of these potential complications.

Table 2: Common and Serious Adverse Reactions to Lansoprazole

Adverse Reaction CategorySpecific Adverse ReactionReported Incidence/Frequency (Source)Key Clinical Considerations/Notes
CommonDiarrhea3.8% (lansoprazole) vs 2.3% (placebo) in short-term trials 12Most common; incidence may be dose-related.
Abdominal Pain2.1% (lansoprazole) vs 1.2% (placebo) 12
Nausea1.3% (lansoprazole) vs 1.2% (placebo) 12
Constipation1.0% (lansoprazole) vs 0.4% (placebo) 12
Headache>1% (more common on placebo in some studies) 12
SeriousHypersensitivity Reactions (Anaphylaxis, Angioedema)Rare 1Contraindicated if known severe hypersensitivity. Discontinue immediately.
Severe Cutaneous Adverse Reactions (SJS, TEN)Rare 1Discontinue at first sign of SCARs. Potentially life-threatening.
Acute Interstitial Nephritis (AIN/ATIN)Rare 1May occur at any time; discontinue lansoprazole if AIN develops.
HepatotoxicityRare, mostly mild 7Usually hepatocellular or mixed pattern; DRESS syndrome reported.
Long-Term RisksBone Fractures (hip, wrist, spine)Associated with long-term (≥1 year) and/or high-dose use 12Use lowest dose/shortest duration. Consider osteoporosis risk factors.
HypomagnesemiaRare; with prolonged use (≥3 months, mostly >1 year) 12Monitor magnesium levels periodically in at-risk patients. May require magnesium supplementation and/or PPI discontinuation.
Clostridioides difficile-Associated DiarrheaIncreased risk, especially in hospitalized patients 12Consider CDAD if diarrhea persists. Use lowest dose/shortest duration.
Cyanocobalamin (Vitamin B12) DeficiencyAssociated with long-term use (e.g., >3 years) 28Consider in patients with relevant clinical symptoms. Due to hypo- or achlorhydria.
Fundic Gland PolypsAssociated with long-term use 10Generally benign; may regress on discontinuation. Linked to hypergastrinemia.
Cutaneous and Systemic Lupus ErythematosusRare; new onset or exacerbation 7Discontinue PPI and refer to specialist.

SJS = Stevens-Johnson syndrome; TEN = Toxic Epidermal Necrolysis; ATIN = Acute Tubulointerstitial Nephritis.

6. Drug Interactions

Lansoprazole is subject to several clinically significant drug interactions, which can arise from its effect on gastric pH, its metabolism via the cytochrome P450 system, or other mechanisms. Careful review of concomitant medications is essential when prescribing lansoprazole.

6.1. Drugs with pH-Dependent Absorption

By increasing intragastric pH, lansoprazole can alter the absorption of drugs whose solubility and absorption are pH-dependent. This is a class effect common to all acid-suppressing agents.

  • Drugs with Decreased Absorption (Reduced Efficacy):
  • Antifungal agents: The absorption of ketoconazole and itraconazole is significantly reduced when co-administered with lansoprazole, potentially leading to therapeutic failure.[1]
  • Atazanavir: Lansoprazole can substantially reduce the plasma concentrations of atazanavir, an antiretroviral agent, by decreasing its absorption. Co-administration is generally not recommended.[18]
  • Iron salts and ampicillin esters: Absorption of these agents may also be reduced.[1]
  • Drugs with Potentially Increased Absorption:
  • Digoxin: Lansoprazole may increase the plasma concentration of digoxin, potentially increasing the risk of digoxin toxicity. Monitoring of digoxin levels may be necessary.[1]

The clinical impact of these interactions depends on the magnitude of the absorption change and the therapeutic index of the co-administered drug. For drugs whose absorption is critically dependent on an acidic environment, alternative therapeutic strategies or careful monitoring may be required.

6.2. Interactions via Metabolic Pathways (CYP Enzymes)

Lansoprazole is primarily metabolized by CYP2C19 and, to a lesser extent, by CYP3A4.[1] This metabolic profile creates potential for interactions with other drugs that are substrates, inhibitors, or inducers of these enzymes.

  • Warfarin: While pharmacokinetic studies in healthy subjects indicated that single or multiple 60 mg doses of lansoprazole did not significantly affect warfarin pharmacokinetics or prothrombin time [18], postmarketing reports have described increased International Normalized Ratio (INR) and prothrombin time in patients receiving PPIs (including lansoprazole) and warfarin concomitantly. This may lead to an increased risk of abnormal bleeding. Therefore, patients treated concomitantly with lansoprazole and warfarin may require monitoring for increases in INR and prothrombin time.[12] The interaction is generally classified as minor but warrants cautious management.
  • Tacrolimus: Co-administration of lansoprazole and tacrolimus may significantly increase whole blood concentrations of tacrolimus. This interaction is particularly relevant in transplant patients who are intermediate or poor metabolizers of CYP2C19, as lansoprazole can inhibit the CYP3A4-mediated metabolism of tacrolimus in these individuals.[12] Increased tacrolimus levels can heighten the risk of serious side effects, including nephrotoxicity, neurotoxicity, diabetes, and hyperkalemia. Dose adjustment of tacrolimus or more frequent monitoring of its blood levels may be necessary.[60]
  • Theophylline: Lansoprazole may cause a small (approximately 10%), probably not clinically significant, increase in the clearance of theophylline. However, titration of theophylline dosage may be required in some individuals to maintain therapeutic concentrations.[12]
  • Cilostazol: Lansoprazole has the potential to increase the plasma concentrations of cilostazol, thereby increasing the risk of cilostazol-related toxicity.[1]
  • Diazepam: Some general PPI information suggests potential for prolonged excretion of diazepam.[1] However, the FDA-approved lansoprazole label states that lansoprazole does not have clinically significant interactions with diazepam.[18] This minor discrepancy suggests that any interaction is likely not of major clinical concern for most patients.

6.3. Other Notable Interactions

  • Clopidogrel: The interaction between PPIs and clopidogrel has been a subject of considerable discussion due to clopidogrel's activation by CYP2C19.
  • The FDA-approved prescribing information for lansoprazole states that concomitant administration of lansoprazole and clopidogrel in healthy subjects had no clinically important effect on exposure to the active metabolite of clopidogrel or on clopidogrel-induced platelet inhibition. Consequently, no dose adjustment of clopidogrel is deemed necessary when administered with an approved dose of lansoprazole.[12]
  • However, broader concerns exist regarding CYP2C19-inhibiting PPIs (particularly omeprazole and esomeprazole) reducing clopidogrel's antiplatelet effect.[61] Lansoprazole 30 mg daily does not appear to significantly reduce overall clopidogrel exposure, although high-dose lansoprazole (60 mg daily) might cause a modest reduction (18%) in the Cmax of clopidogrel's active metabolite.[62]
  • Regulatory agencies like the MHRA and EMA advise against the concurrent use of clopidogrel with omeprazole and esomeprazole but do not extend this strong recommendation to other PPIs such as lansoprazole.[62]
  • The current understanding suggests that lansoprazole is a relatively safer PPI choice compared to omeprazole or esomeprazole when co-administration with clopidogrel is necessary. However, the potential for an interaction, especially with higher lansoprazole doses or in patients who are poor metabolizers of CYP2C19 (affecting clopidogrel activation), should be kept in mind.
  • Methotrexate: Concomitant use of PPIs with methotrexate, primarily when methotrexate is administered at high doses, may elevate and prolong serum concentrations of methotrexate and/or its metabolite, 7-hydroxymethotrexate. This can potentially lead to methotrexate-related toxicities.[12] The proposed mechanism involves PPI inhibition of active tubular secretion of methotrexate and its metabolite via renal H+/K+ ATPase pumps, or inhibition of the breast cancer resistance protein (BCRP)-mediated transport.[63] For patients receiving high-dose methotrexate, temporary withdrawal of the PPI may be considered.[12]
  • Sucralfate: Sucralfate can delay the absorption and reduce the bioavailability of lansoprazole by 17-30%. To minimize this interaction, lansoprazole should be administered at least 30 minutes prior to sucralfate.[10]
  • Antacids: Co-administration of antacids does not appear to have a clinically significant impact on the efficacy of lansoprazole.[18]

The diverse nature of these interactions, affecting drug absorption, metabolism, and transport, necessitates a thorough medication review for any patient starting or continuing lansoprazole therapy.

Table 3: Clinically Significant Drug Interactions with Lansoprazole

Interacting Drug/ClassPotential Effect on Interacting Drug or LansoprazoleMechanism (if known)Clinical Significance/RecommendationReference(s)
Drugs with pH-Dependent Absorption
Ketoconazole, ItraconazoleDecreased absorption and efficacy of antifungalIncreased gastric pH reduces solubility/dissolutionAvoid concomitant use or monitor for reduced antifungal efficacy; consider alternative antifungals.1
AtazanavirSignificantly reduced plasma concentrations and efficacy of atazanavirIncreased gastric pH reduces absorptionCo-administration not recommended.18
DigoxinPotentially increased plasma concentrations of digoxinIncreased gastric pH may enhance digoxin absorptionMonitor digoxin levels and for signs of toxicity if co-administered.1
Drugs Interacting via Metabolic Pathways
WarfarinPotential for increased INR and prothrombin time; risk of bleedingUncertain; postmarketing reports suggest interaction despite no significant PK effect in studies.Monitor INR and prothrombin time frequently, especially upon initiation or discontinuation of lansoprazole. Adjust warfarin dose as needed.12
TacrolimusIncreased whole blood levels of tacrolimus, risk of toxicityInhibition of CYP3A4-mediated tacrolimus metabolism by lansoprazole, especially in CYP2C19 IM/PMs.Monitor tacrolimus blood levels closely, especially in CYP2C19 IM/PMs. Dose adjustment of tacrolimus may be necessary.12
TheophyllineSmall (approx. 10%) increase in theophylline clearanceUncertainProbably not clinically significant for most; titration of theophylline dosage may be required in some individuals.12
Other Notable Interactions
ClopidogrelStandard dose lansoprazole: No clinically important effect on active metabolite/platelet inhibition. High dose lansoprazole: Potential minor reduction in Cmax of active metabolite.Lansoprazole is a weaker inhibitor of CYP2C19 compared to omeprazole/esomeprazole.Standard dose lansoprazole generally considered acceptable with clopidogrel. Avoid high doses if concern exists. Preferable to omeprazole/esomeprazole.12
Methotrexate (especially high dose)Elevated and prolonged serum levels of methotrexate and/or its metabolite; risk of toxicityInhibition of renal H+/K+ ATPase pumps or BCRP transport by PPIs, reducing methotrexate elimination.Consider temporary withdrawal of lansoprazole during high-dose methotrexate therapy. Monitor for methotrexate toxicity.12
SucralfateDelayed absorption and reduced bioavailability of lansoprazole (by 17-30%)Physical binding or altered dissolution environment.Administer lansoprazole at least 30 minutes before sucralfate.10

INR = International Normalized Ratio; PK = Pharmacokinetics; IM = Intermediate Metabolizer; PM = Poor Metabolizer; BCRP = Breast Cancer Resistance Protein.

7. Use in Specific Populations

The use of lansoprazole in specific patient populations, such as pregnant or lactating women, children, the elderly, and those with hepatic impairment, requires careful consideration of potential risks and benefits, as well as possible dosage adjustments.

7.1. Pregnancy

  • FDA Pregnancy Category and Risk Summary: Historically, lansoprazole was classified as FDA Pregnancy Category B, indicating that animal reproduction studies did not demonstrate a fetal risk, and there were no adequate and well-controlled studies in pregnant women, or animal studies showed an adverse effect not confirmed in controlled human studies.[65] Omeprazole was Category C. The FDA has since transitioned away from these letter categories to a narrative risk summary. Current FDA labeling for lansoprazole states that observational studies do not suggest an association with adverse developmental outcomes; however, due to methodological limitations, drug-associated risks cannot be definitively established or excluded. Animal studies have shown fetal mortality and lower fetal weight at high doses, but no evidence of teratogenicity.[66]
  • Safety Data: A substantial body of evidence from human observational studies and meta-analyses suggests that PPIs, including lansoprazole and the more extensively studied omeprazole, are generally not associated with a significant increase in the risk of major congenital malformations, spontaneous abortions, or premature deliveries when used during pregnancy.[65] A large Danish cohort study published in 2010, involving over 840,000 births, found no link between first-trimester PPI use (with omeprazole being the most common) and birth defects.[65]
  • Pre-conception Use: An interesting finding from the 2010 Danish study was a suggestion of an increased risk of birth defects in women who used PPIs 1 to 4 weeks before conception. However, this finding requires further investigation and was not consistent across all analyses (e.g., for omeprazole alone or OTC PPIs).[65] Some gastroenterologists suggest that patients with GERD might consider discontinuing PPI therapy when planning pregnancy, if clinically feasible, pending more definitive data.[65]
  • Guideline Considerations for GERD/Heartburn in Pregnancy (including ACOG perspectives):
  • The American College of Obstetricians and Gynecologists (ACOG) and other guidelines generally recommend a step-up approach for managing GERD and heartburn during pregnancy.[68]
  • Lifestyle modifications (e.g., dietary changes, avoiding trigger foods, elevating the head of the bed) are the initial recommended strategy.[67]
  • If lifestyle changes are insufficient, antacids containing calcium carbonate or aluminum hydroxide (e.g., Tums, Maalox) are considered first-line medical therapy due to their safety profile.[65] Sucralfate can also be used.
  • If symptoms persist, H2-receptor antagonists (e.g., famotidine, ranitidine [though ranitidine availability has changed due to unrelated safety concerns]) are generally considered safe and can be used next.[65]
  • Proton Pump Inhibitors (PPIs), including lansoprazole and omeprazole, are typically reserved for women with intractable symptoms or complicated reflux disease (e.g., erosive esophagitis), particularly in the second and third trimesters.[65] Some sources suggest lansoprazole may be a preferred PPI due to its animal and human pregnancy safety profile [68], while others favor omeprazole due to the larger volume of available safety data.[67] Ultimately, the choice of PPI should be individualized. Despite the historical FDA category differences, current evidence generally supports the safety of lansoprazole and other PPIs during pregnancy when clinically indicated, especially after the first trimester. The pre-conception risk signal warrants further research. A conservative, step-wise approach to managing GERD in pregnancy is consistently advised.

7.2. Lactation

The use of lansoprazole during breastfeeding requires careful consideration due to limited direct human data.

  • Excretion in Breast Milk: There is no specific published information available on the excretion of lansoprazole into human breast milk.[8] Lansoprazole is known to be excreted into animal milk.[66]
  • Potential Effects on Nursing Infant: The effects of lansoprazole exposure via breast milk on the nursing infant are unknown.[66] However, lansoprazole has been used directly and safely in newborn infants for therapeutic purposes, which suggests that the small amounts potentially ingested through breast milk are unlikely to be harmful.[8] Nevertheless, a theoretical concern exists due to findings of tumorigenicity in animal carcinogenicity studies with lansoprazole, raising a potential risk of serious adverse reactions in the nursing infant if significant exposure were to occur.[66]
  • Guideline Considerations and Expert Opinions:
  • UKTIS/SPS (UK Teratology Information Service / Specialist Pharmacy Service): While any PPI can be used during breastfeeding, omeprazole and pantoprazole are often preferred choices because there is evidence they are excreted into breast milk in very small amounts, and they have more supporting data for use. It is generally believed that any PPI that passes into breast milk is likely to be degraded by the acidic environment of the infant's gastrointestinal tract, limiting systemic absorption by the infant. Lansoprazole can be used with caution, but omeprazole or pantoprazole are preferred. Although there is no published evidence of lansoprazole use during breastfeeding, it is not expected to pass into breast milk in significant amounts, and side effects in breastfed infants are therefore not anticipated.[69]
  • LactMed® (Drugs and Lactation Database - NCBI): LactMed also notes the lack of information on lansoprazole in breast milk but concurs that since lansoprazole is used safely in newborns, amounts in milk are unlikely to be harmful. It lists antacids, cimetidine, famotidine, omeprazole, pantoprazole, and sucralfate as alternative drugs to consider.[8]
  • Midlands Medicines Information Service (NHS): Similar to other sources, this service notes the absence of human data for lansoprazole in breast milk. Due to its structural and pharmacokinetic similarity to omeprazole (for which some data exist showing low milk levels), amounts of lansoprazole in breast milk are thought to be small and likely destroyed in the infant's stomach. They suggest that use could proceed with caution if a PPI is necessary.[70]
  • Effects on Lactation (Maternal Prolactin): There have been reports of gynecomastia and galactorrhea associated with lansoprazole use in some individuals (both male and female), possibly due to hyperprolactinemia.[8] However, the clinical significance of this on milk production in women with established lactation is unknown and likely not significant.[69]

Given the absence of direct human milk data for lansoprazole, a cautious approach is generally advised. If a PPI is essential during lactation, agents with more established safety data in breastfeeding, such as omeprazole or pantoprazole, are often preferred. The acid lability of PPIs is a key factor suggesting that infant systemic exposure via breast milk would likely be minimal.

7.3. Pediatric Use

The use of lansoprazole in pediatric populations has been established for specific indications and age groups, with pharmacokinetic profiles and dosing regimens studied (details covered in Sections 2.3 Pharmacokinetics and 4.3 Pediatric Dosing).

  • Efficacy and Safety: Lansoprazole is considered safe and effective for the short-term treatment of symptomatic GERD and erosive esophagitis in children aged 1 to 17 years.[71]
  • Infants (<1 year): Safety and efficacy have not been established in children younger than 1 year of age.[71] There is a potential concern that lansoprazole use in this age group may increase the risk for heart problems, such as heart valve thickening.[71] As noted previously, the manufacturer did not pursue an indication for GERD in infants due to a lack of demonstrated efficacy in clinical trials.[24]

7.4. Geriatric Use

Clinical studies of lansoprazole did not show overall differences in safety or effectiveness when comparing elderly subjects to younger subjects. However, it is acknowledged that greater sensitivity in some older individuals cannot be entirely ruled out.[71] Elderly patients are also more likely to have pre-existing renal or hepatic impairment, which may necessitate caution or dosage adjustments.[27]

7.5. Hepatic Impairment

Lansoprazole is extensively metabolized in the liver. Therefore, hepatic impairment can significantly affect its pharmacokinetics.

  • Mild (Child-Pugh Class A) or Moderate (Child-Pugh Class B) Hepatic Impairment: No dosage adjustment is typically necessary.[29]
  • Severe (Child-Pugh Class C) Hepatic Impairment: A dose reduction is recommended. The FDA prescribing information suggests considering a maximum daily dose of 15 mg for patients with severe liver disease.[12] This is due to the prolonged half-life and increased exposure observed in this population.[13]

8. Pharmacogenomics: CYP2C19 Polymorphisms

The metabolism of lansoprazole is significantly influenced by genetic variations in the CYP2C19 gene, which encodes a key enzyme in its clearance pathway. These polymorphisms can lead to distinct metabolizer phenotypes, impacting drug exposure and clinical outcomes.

  • Impact of CYP2C19 Metabolizer Status on Lansoprazole Pharmacokinetics: Lansoprazole is primarily metabolized by CYP2C19, with CYP3A4 playing a lesser role.1 Genetic polymorphisms in CYP2C19 result in different enzyme activity levels, leading to variations in lansoprazole plasma concentrations and Area Under the Curve (AUC).20 The effect of CYP2C19 polymorphism on individual PPIs varies, with omeprazole being most affected, followed by lansoprazole, dexlansoprazole, pantoprazole, esomeprazole, and rabeprazole having minimal CYP2C19 involvement.72
  • Poor Metabolizers (PMs): Individuals with two no-function CYP2C19 alleles (e.g., *2/*2, *2/*3, *3/*3) exhibit significantly reduced enzyme activity. This leads to decreased clearance of lansoprazole and consequently higher plasma concentrations and AUC.[22] PMs may experience enhanced therapeutic effects but could also be at a theoretically increased risk of concentration-dependent adverse drug reactions (ADRs), although this link to ADRs is not consistently established for all PPIs.[72]
  • Intermediate Metabolizers (IMs): Individuals carrying one normal or increased function allele and one no-function allele (e.g., *1/*2, *1/*3, *2/*17) or two decreased function alleles have reduced enzyme activity compared to normal metabolizers. Their exposure to lansoprazole is generally between that of PMs and NMs.[22]
  • Normal Metabolizers (NMs) (formerly Extensive Metabolizers - EMs): Individuals with two normal function alleles (e.g., *1/*1) have standard enzyme activity and typical lansoprazole metabolism.[22] They may experience lower efficacy for certain indications compared to PMs or IMs when standard doses are used.
  • Rapid Metabolizers (RMs): Individuals carrying one normal function allele and one increased function allele (e.g., *1/*17) exhibit increased enzyme activity, leading to faster metabolism of lansoprazole, lower plasma concentrations, and reduced AUC compared to NMs.[72] This can increase the risk of therapeutic failure.
  • Ultrarapid Metabolizers (UMs): Individuals with two increased function alleles (e.g., *17/*17) or combinations leading to very high enzyme activity metabolize lansoprazole very rapidly, resulting in significantly lower plasma exposure and a higher likelihood of treatment failure with standard doses.[22]
  • Clinical Implications of CYP2C19 Polymorphisms:
  • Helicobacter pylori Eradication: The efficacy of standard triple therapy regimens (PPI, amoxicillin, clarithromycin) for H. pylori eradication is significantly influenced by the patient's CYP2C19 genotype. UMs and RMs often exhibit lower eradication rates with standard PPI doses due to insufficient acid suppression, which can affect antibiotic stability and efficacy.[74] Conversely, PMs and IMs tend to have higher eradication rates.
  • Gastroesophageal Reflux Disease (GERD) / Erosive Esophagitis (EE): NMs, RMs, and UMs may be at an increased risk of refractory GERD or relapse of EE when treated with standard lansoprazole doses, compared to PMs and IMs who achieve higher intragastric pH levels.[74] For instance, studies have shown lower healing rates of EE and earlier symptomatic recurrence during maintenance therapy in NMs compared to PMs on standard lansoprazole doses.[76]
  • Dosing Recommendations Based on CYP2C19 Genotype: Both the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) have issued guidelines for PPI dosing based on CYP2C19 genotype, although some differences exist in their specific recommendations.
  • CPIC Guidelines (2020) for Lansoprazole [74]:
  • CYP2C19 Ultrarapid Metabolizer (UM): Increase starting daily dose by 100%. The daily dose may be given in divided doses. Monitor for efficacy. (Strength of recommendation: Optional).
  • CYP2C19 Rapid Metabolizer (RM): Initiate standard starting daily dose. For the treatment of H. pylori infection and erosive esophagitis, consider increasing the dose by 50-100%. The daily dose may be given in divided doses. Monitor for efficacy. (Strength: Moderate).
  • CYP2C19 Normal Metabolizer (NM): Initiate standard starting daily dose. For the treatment of H. pylori infection and erosive esophagitis, consider increasing the dose by 50-100%. The daily dose may be given in divided doses. Monitor for efficacy. (Strength: Moderate).
  • CYP2C19 Intermediate Metabolizer (IM): Initiate standard starting daily dose. For chronic therapy (>12 weeks), if efficacy is achieved, consider a 50% reduction in the daily dose. Monitor for continued efficacy. (Strength: Optional).
  • CYP2C19 Poor Metabolizer (PM): Initiate standard starting daily dose. For chronic therapy (>12 weeks), if efficacy is achieved, consider a 50% reduction in the daily dose. Monitor for continued efficacy. (Strength: Moderate).
  • Dutch Pharmacogenetics Working Group (DPWG) for Lansoprazole [22]:
  • CYP2C19 Ultrarapid Metabolizer (UM): For H. pylori eradication, use a 200% increased dose.[22] For other indications, be alert to reduced effectiveness and, if necessary, use a similarly increased dose (200% or 4-fold depending on source interpretation).[22]
  • CYP2C19 Poor Metabolizer (PM) & Intermediate Metabolizer (IM): No specific initial therapeutic dose adjustment is recommended, but it is noted that these phenotypes experience increased therapeutic efficacy for reflux esophagitis and H. pylori eradication with standard doses.[22] The DPWG guidelines generally do not recommend dose reductions for PMs/IMs due to a lack of consistent correlation between increased plasma exposure and increased ADRs for PPIs.[72]

The differing recommendations between CPIC and DPWG, particularly for PMs and IMs, highlight the evolving nature of pharmacogenomic guidance and the complexities in translating pharmacokinetic differences into definitive clinical actions, especially concerning long-term therapy and adverse events. However, there is a stronger consensus on the need for dose increases in UMs and RMs for indications where profound acid suppression is critical. The potential for genotype-guided dosing to optimize lansoprazole therapy, particularly in challenging cases like H. pylori eradication or refractory GERD, is significant. It allows for a more personalized approach, aiming to maximize efficacy in rapid metabolizers and potentially minimize long-term exposure or cost in poor metabolizers once efficacy is achieved.

Table 4: CYP2C19 Genotype-Based Dosing Recommendations for Lansoprazole (Summary from CPIC Guidelines 2020)

CYP2C19 PhenotypeImplication for Lansoprazole Metabolism/ExposureCPIC Therapeutic Recommendation for LansoprazoleStrength of RecommendationReference(s)
Ultrarapid Metabolizer (UM)Significantly decreased plasma concentrations; increased risk of therapeutic failure.Increase starting daily dose by 100%. Daily dose may be given in divided doses. Monitor for efficacy.Optional74
Rapid Metabolizer (RM)Decreased plasma concentrations compared to NMs; increased risk of therapeutic failure.Initiate standard starting daily dose. For H. pylori infection and erosive esophagitis, consider increasing dose by 50-100%. Daily dose may be given in divided doses. Monitor for efficacy.Moderate74
Normal Metabolizer (NM)Normal PPI metabolism; may be at increased risk of therapeutic failure compared to IMs and PMs.Initiate standard starting daily dose. For H. pylori infection and erosive esophagitis, consider increasing dose by 50-100%. Daily dose may be given in divided doses. Monitor for efficacy.Moderate74
Intermediate Metabolizer (IM)Increased plasma concentrations compared to NMs; increased chance of efficacy.Initiate standard starting daily dose. For chronic therapy (>12 weeks) and efficacy achieved, consider 50% reduction in daily dose. Monitor for continued efficacy.Optional74
Poor Metabolizer (PM)Significantly increased plasma concentrations compared to NMs; increased chance of efficacy and potentially toxicity (though ADR link is less clear).Initiate standard starting daily dose. For chronic therapy (>12 weeks) and efficacy achieved, consider 50% reduction in daily dose. Monitor for continued efficacy.Moderate74

CPIC = Clinical Pharmacogenetics Implementation Consortium. Recommendations are based on the 2020 CPIC guideline for CYP2C19 and Proton Pump Inhibitors.

9. Conclusion and Clinical Considerations

Lansoprazole is a proton pump inhibitor with a long history of clinical use and proven efficacy in the management of a wide spectrum of acid-related gastrointestinal disorders in both adult and pediatric populations. Its mechanism of action, involving irreversible inhibition of the gastric H+/K+-ATPase, leads to potent and sustained suppression of gastric acid secretion, which is fundamental to its therapeutic effects in conditions such as peptic ulcer disease, GERD, erosive esophagitis, and Zollinger-Ellison syndrome, as well as its role in H. pylori eradication regimens.

The clinical efficacy of lansoprazole is generally comparable to that of other PPIs. However, subtle differences may exist in terms of potency of acid suppression, speed of symptom relief, or efficacy in specific patient subgroups or disease severities when compared directly with agents like esomeprazole or the newer class of P-CABs. Such nuances, alongside cost-effectiveness and formulary availability, may influence the choice of agent in clinical practice.

Optimal use of lansoprazole necessitates careful attention to several key factors. Correct administration timing, specifically before meals, is critical due to the significant impact of food on its bioavailability. While generally well-tolerated for short-term use, the recognized long-term risks associated with PPI therapy—including an increased risk of bone fractures, hypomagnesemia, Clostridioides difficile-associated diarrhea, and potential vitamin B12 deficiency—mandate a cautious approach to chronic use. This involves regularly reassessing the need for continued therapy and employing the lowest effective dose for the shortest duration appropriate to the clinical condition.

The pharmacogenomic impact of CYP2C19 genetic polymorphisms on lansoprazole metabolism and clinical response is well-documented. Individuals with ultrarapid or rapid metabolizer phenotypes may experience therapeutic failure with standard doses, particularly for indications requiring profound acid suppression like H. pylori eradication or severe erosive esophagitis. Conversely, poor metabolizers may achieve therapeutic goals with standard or even reduced doses for long-term maintenance, although the link between higher exposure in PMs and an increased risk of adverse drug reactions is not definitively established for all PPI-related ADRs. Genotype-guided dosing, as suggested by guidelines from CPIC and DPWG, offers a pathway towards personalized lansoprazole therapy, potentially improving efficacy and minimizing risks in selected patients.

Furthermore, a thorough understanding and management of potential drug interactions are crucial. Lansoprazole can affect the absorption of pH-dependent drugs and interact with medications metabolized by CYP enzymes, such as warfarin and tacrolimus, or drugs whose efficacy can be modulated by PPIs, like methotrexate and clopidogrel.

The use of lansoprazole in special populations, such as pregnant or lactating women, requires a careful risk-benefit assessment. While current evidence is generally reassuring regarding its safety during pregnancy, particularly after the first trimester, data on its use during lactation are limited, often leading to a preference for other PPIs with more established lactation safety profiles.

In conclusion, lansoprazole remains a cornerstone in the pharmacological management of acid-related disorders. Its effective and sustained acid suppression capabilities are well-established. However, maximizing its therapeutic benefits while minimizing potential risks involves a comprehensive approach that considers the specific clinical indication, patient-specific factors including pharmacogenomic profile and comorbidities, potential drug interactions, and the evolving understanding of the safety implications of long-term PPI use. Judicious prescribing, regular monitoring, and patient education are paramount to the optimal and safe utilization of lansoprazole in clinical practice.

Works cited

  1. Lansoprazole - Wikipedia, accessed May 29, 2025, https://en.wikipedia.org/wiki/Lansoprazole
  2. Lansoprazole - brand name list from Drugs.com, accessed May 29, 2025, https://www.drugs.com/ingredient/lansoprazole.html
  3. my.clevelandclinic.org, accessed May 29, 2025, https://my.clevelandclinic.org/health/drugs/18428-lansoprazole-capsules#:~:text=Lansoprazole%20is%20a%20medication%20that,this%20medication%20is%20Prevacid%C2%AE.
  4. Lansoprazole: Side Effects, Uses, Dosage, Interactions, Warnings, accessed May 29, 2025, https://www.rxlist.com/lansoprazole/generic-drug.htm
  5. Lansoprazole (AG-1749, CAS Number: 103577-45-3) | Cayman Chemical, accessed May 29, 2025, https://www.caymanchem.com/product/13627/lansoprazole
  6. Lansoprazole | CAS 103577-45-3 | AG-1749 - Tocris Bioscience, accessed May 29, 2025, https://www.tocris.com/products/lansoprazole_2582
  7. Lansoprazole - LiverTox - NCBI Bookshelf, accessed May 29, 2025, https://www.ncbi.nlm.nih.gov/books/NBK548452/
  8. Lansoprazole - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed May 29, 2025, https://www.ncbi.nlm.nih.gov/books/NBK501283/
  9. go.drugbank.com, accessed May 29, 2025, https://go.drugbank.com/drugs/DB00448#:~:text=Lansoprazole%20marketed%20under%20the%20brand,classified%20as%20a%20substituted%20benzimidazole.
  10. Lansoprazole: MedlinePlus Drug Information, accessed May 29, 2025, https://medlineplus.gov/druginfo/meds/a695020.html
  11. Discovery and development of proton pump inhibitors - Wikipedia, accessed May 29, 2025, https://en.wikipedia.org/wiki/Discovery_and_development_of_proton_pump_inhibitors
  12. www.accessdata.fda.gov, accessed May 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020406s078-021428s025lbl.pdf
  13. Lansoprazole | Drug Lookup | Pediatric Care Online - AAP Publications, accessed May 29, 2025, https://publications.aap.org/pediatriccare/drug-monograph/18/5714/Lansoprazole
  14. Duration of effect of lansoprazole on gastric pH and acid secretion in normal male subjects, accessed May 29, 2025, https://www.researchgate.net/publication/12193272_Duration_of_effect_of_lansoprazole_on_gastric_pH_and_acid_secretion_in_normal_male_subjects
  15. The comparative effects of lansoprazole, omeprazole, and ranitidine in suppressing gastric acid secretion - Portal de Periódicos da CAPES, accessed May 29, 2025, https://www.periodicos.capes.gov.br/index.php/acervo/buscador.html?task=detalhes&id=W2069810458
  16. Effects on 24-hour intragastric pH: a comparison of lansoprazole administered nasogastrically in apple juice and pantoprazole administered intravenously - PubMed, accessed May 29, 2025, https://pubmed.ncbi.nlm.nih.gov/11467632/
  17. Relative potency of proton-pump inhibitors - Comparison of effects on intragastric pH, accessed May 29, 2025, https://www.researchgate.net/publication/23388932_Relative_potency_of_proton-pump_inhibitors_-_Comparison_of_effects_on_intragastric_pH
  18. Summary Review - accessdata.fda.gov, accessed May 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/022327s000_SumR.pdf
  19. prevacid - accessdata.fda.gov, accessed May 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/020406s067,021281s024,021428s017lbl.pdf
  20. Proton pump inhibitors: from CYP2C19 pharmacogenetics to precision medicine - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5942154/
  21. Annotation of FDA Label for lansoprazole and CYP2C19 - PharmGKB, accessed May 29, 2025, https://www.pharmgkb.org/labelAnnotation/PA166104898
  22. CYP2C19: lansoprazole 2502 to 2504 - G-Standaard, accessed May 29, 2025, https://www.g-standaard.nl/risicoanalyse/M0002502.pdf
  23. Good clinical practice recommendations for proton pump inhibitor prescription and deprescription. A review by experts from the AMG, accessed May 29, 2025, https://www.revistagastroenterologiamexico.org/en-good-clinical-practice-recommendations-for-articulo-S2255534X25000209
  24. PEDIATRIC REVIEW - FDA, accessed May 29, 2025, https://www.fda.gov/media/71457/download
  25. Eradication of Helicobacter Pylori, accessed May 29, 2025, https://www.nottsapc.nhs.uk/media/ywedjlbg/eradication-of-helicobacter-pylori.pdf
  26. What are the latest guideline recommendations for the treatment of Helicobacter pylori infection in the U.S.? | Drug Information Group | University of Illinois Chicago, accessed May 29, 2025, https://dig.pharmacy.uic.edu/faqs/2025-2/january-2025-faqs/what-are-the-latest-guideline-recommendations-for-the-treatment-of-helicobacter-pylori-infection-in-the-u-s/
  27. Lansoprazole, amoxicillin, and clarithromycin (oral route) - Mayo Clinic, accessed May 29, 2025, https://www.mayoclinic.org/drugs-supplements/lansoprazole-amoxicillin-and-clarithromycin-oral-route/description/drg-20137709
  28. This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed May 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/020406s095,021428s042lbl.pdf
  29. Prevacid, Prevacid Solu Tab (lansoprazole) dosing, indications, interactions, adverse effects, and more - Medscape, accessed May 29, 2025, https://reference.medscape.com/drug/prevacid-solu-tab-lansoprazole-341991
  30. Lansoprazole: Pediatric drug information, accessed May 29, 2025, https://doctorabad.com/uptodate/d/topic.htm?path=lansoprazole-pediatric-drug-information
  31. Comparative effectiveness of proton pump inhibitors - Therapeutics Letter - NCBI Bookshelf, accessed May 29, 2025, https://www.ncbi.nlm.nih.gov/books/NBK598449/
  32. Clinical Briefs - AAFP, accessed May 29, 2025, https://www.aafp.org/pubs/afp/issues/2003/0915/p1224.html
  33. Choosing One PPI Treatment Over Another - AAFP, accessed May 29, 2025, https://www.aafp.org/pubs/afp/issues/2007/1101/p1273.html
  34. Omeprazole vs Lansoprazole in the Management of Gastroesophageal Reflux Disease: A Systematic Literature Review | Journal of Medical Research and Innovation, accessed May 29, 2025, https://jmrionline.com/jmri/article/view/204
  35. Comparative effectiveness of different recommended doses of omeprazole and lansoprazole for gastroesophageal reflux disease - e-Century Publishing Corporation, accessed May 29, 2025, https://e-century.us/files/ijcep/10/4/ijcep0048253.pdf
  36. Comparison of four proton pump inhibitors for the short-term treatment of esophagitis in elderly patients - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4611579/
  37. Comparison of four proton pump inhibitors for the short-term treatment of esophagitis in elderly patients - Baishideng Publishing Group, accessed May 29, 2025, https://www.wjgnet.com/1007-9327/full/v13/i33/4467.htm
  38. Comparative effectiveness and acceptability of the FDA-licensed proton pump inhibitors for erosive esophagitis: A PRISMA-compliant network meta-analysis - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5626283/
  39. (PDF) Effectiveness and Tolerability of Different Recommended Doses of PPIs and H2RAs in GERD: Network Meta-Analysis and GRADE system - ResearchGate, accessed May 29, 2025, https://www.researchgate.net/publication/312567512_Effectiveness_and_Tolerability_of_Different_Recommended_Doses_of_PPIs_and_H2RAs_in_GERD_Network_Meta-Analysis_and_GRADE_system
  40. Esomeprazole (40 Mg) Compared With Lansoprazole (30 Mg) in The Treatment of Erosive Esophagitis | Request PDF - ResearchGate, accessed May 29, 2025, https://www.researchgate.net/publication/11441823_Esomeprazole_40_mg_compared_with_lansoprazole_30_mg_in_the_treatment_of_erosive_esophagitis
  41. A Deep Dive Into the 2022 ACG GERD Guideline Update - HCPLive, accessed May 29, 2025, https://www.hcplive.com/view/deep-dive-2022-acg-gerd-guideline-update
  42. Meta-analysis of the efficacy of lansoprazole and omeprazole for the treatment of H.pylori-associated duodenal ulcer - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4697672/
  43. (PDF) Comparative efficiency and safety of potassium competitive acid blockers versus Lansoprazole in peptic ulcer: a systematic review and meta-analysis - ResearchGate, accessed May 29, 2025, https://www.researchgate.net/publication/377722476_Comparative_efficiency_and_safety_of_potassium_competitive_acid_blockers_versus_Lansoprazole_in_peptic_ulcer_a_systematic_review_and_meta-analysis
  44. Comparative efficiency and safety of potassium competitive acid blockers versus Lansoprazole in peptic ulcer: a systematic review and meta-analysis - PubMed, accessed May 29, 2025, https://pubmed.ncbi.nlm.nih.gov/38273830/
  45. Comparative efficiency and safety of potassium competitive acid blockers versus Lansoprazole in peptic ulcer: a systematic review and meta-analysis - Frontiers, accessed May 29, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2023.1304552/full
  46. Comparative efficiency and safety of potassium competitive acid blockers versus Lansoprazole in peptic ulcer: a systematic review and meta-analysis, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10808560/
  47. Efficacy and safety of vonoprazan versus proton pump inhibitors in the treatment of peptic ulcer disease: a systematic review and network meta-analysis for randomized controlled trails - Frontiers, accessed May 29, 2025, https://www.frontiersin.org/journals/nutrition/articles/10.3389/fnut.2024.1436993/full
  48. Are proton pump inhibitors the first choice for acute treatment of gastric ulcers? A meta analysis of randomized clinical trials - PubMed Central, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC117603/
  49. Standard-Dose Proton Pump Inhibitors in the Initial Non-eradication Treatment of Duodenal Ulcer - Frontiers, accessed May 29, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2018.01512/full
  50. PREVACID(lansoprazole) Label - accessdata.fda.gov, accessed May 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/021428s028lbl020406s081lbl.pdf
  51. Proton Pump Inhibitors and Fracture Risk: A Review of Current Evidence and Mechanisms Involved - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6540255/
  52. Proton Pump Inhibitors and Fractures in Adults: A Critical Appraisal and Review of the Literature - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7822697/
  53. The possible risks of proton pump inhibitors - The Medical Journal of Australia, accessed May 29, 2025, https://www.mja.com.au/system/files/issues/205_07/10.5694mja16.00750.pdf
  54. Mechanisms of proton pump inhibitor‐induced hypomagnesemia - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9539870/
  55. Proton-pump inhibitor use amongst patients with severe hypomagnesemia - Frontiers, accessed May 29, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2023.1092476/full
  56. Examination of Potential Mechanisms To Explain the Association between Proton Pump Inhibitors and Clostridium difficile Infection - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2764230/
  57. Proton Pump Inhibitors and Clostridium Difficile Infection: Are We Propagating an Already Rapidly Growing Healthcare Problem? - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5051091/
  58. Proton Pump Inhibitors, H2-Receptor Antagonists, Metformin, and Vitamin B-12 Deficiency: Clinical Implications - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6054240/
  59. Lansoprazole and warfarin Interactions - Drugs.com, accessed May 29, 2025, https://www.drugs.com/drug-interactions/lansoprazole-with-warfarin-1433-0-2311-0.html
  60. Lansoprazole and tacrolimus Interactions - Drugs.com, accessed May 29, 2025, https://www.drugs.com/drug-interactions/lansoprazole-with-tacrolimus-1433-0-2142-0.html
  61. Drug Interactions between clopidogrel and lansoprazole - Drugs.com, accessed May 29, 2025, https://www.drugs.com/drug-interactions/clopidogrel-with-lansoprazole-705-0-1433-0.html
  62. Using clopidogrel with proton pump inhibitors (PPIs) – SPS - Specialist Pharmacy Service, accessed May 29, 2025, https://www.sps.nhs.uk/articles/using-clopidogrel-with-proton-pump-inhibitors-ppis/
  63. Drug Interaction Report - methotrexate lansoprazole - Drugs.com, accessed May 29, 2025, https://www.drugs.com/interactions-check.php?drug_list=1433-0,1590-0&professional=1
  64. Drug Interactions between lansoprazole and methotrexate - Drugs.com, accessed May 29, 2025, https://www.drugs.com/drug-interactions/lansoprazole-with-methotrexate-1433-0-1590-0.html
  65. Treatment of Gastroesophageal Reflux Disease During Pregnancy - PMC, accessed May 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3966174/
  66. Lansoprazole Use During Pregnancy - Drugs.com, accessed May 29, 2025, https://www.drugs.com/pregnancy/lansoprazole.html
  67. Proton pump inhibitors (PPIs) - Bumps, accessed May 29, 2025, https://www.medicinesinpregnancy.org/leaflets-a-z/proton-pump-inhibitors/
  68. What are the treatment options for Gastroesophageal Reflux Disease (GERD) during pregnancy? - Dr.Oracle AI, accessed May 29, 2025, https://www.droracle.ai/articles/54723/treatment-of-gerd-in-pregnancy-
  69. Treating heartburn and dyspepsia during breastfeeding – SPS - Specialist Pharmacy Service – The first stop for professional medicines advice, accessed May 29, 2025, https://www.sps.nhs.uk/articles/treating-heartburn-and-dyspepsia-during-breastfeeding/
  70. What is the preferred proton-pump-inhibitor for treatment of reflux in a breast-feeding mother - Midlands Medicines, accessed May 29, 2025, http://www.midlandsmedicines.nhs.uk/filestore/QA98_PPIs_BM.pdf
  71. Lansoprazole (oral route) - Mayo Clinic, accessed May 29, 2025, https://www.mayoclinic.org/drugs-supplements/lansoprazole-oral-route/description/drg-20067214
  72. Association between CYP2C19 polymorphism and proton pump inhibitors adverse drug reactions: a narrative review - Frontiers, accessed May 29, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2025.1523399/full
  73. Association between CYP2C19 polymorphism and proton pump inhibitors adverse drug reactions: a narrative review - Frontiers, accessed May 29, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2025.1523399/pdf
  74. Clinical Annotation for CYP2C19*1, CYP2C19*2, CYP2C19*3; lansoprazole; Gastroesophageal Reflux and Helicobacter Infections (level 1A Efficacy) - PharmGKB, accessed May 29, 2025, https://www.pharmgkb.org/clinicalAnnotation/1183629796
  75. Annotation of CPIC Guideline for lansoprazole, omeprazole, pantoprazole and CYP2C19, accessed May 29, 2025, https://www.pharmgkb.org/guidelineAnnotation/PA166219103
  76. CYP2C19-Proton Pump Inhibitors - CPIC, accessed May 29, 2025, https://cpicpgx.org/wp-content/uploads/2018/02/CYP2C19-PPIs_Lit-Review_CPIC_Handout_02012018.pdf

Published at: May 29, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath