Biaxin, Biaxin Bid, Omeclamox, Prevpac, Voquezna 14 Day Triplepak 20;500;500
Small Molecule
C38H69NO13
81103-11-9
Acute Bacterial Exacerbation of Chronic Bronchitis (ABECB), Acute maxillary sinusitis, Bacterial Infections, Bartonellosis, Community Acquired Pneumonia (CAP), Duodenal ulcer caused by helicobacter pylori, Helicobacter Pylori Infection, Infective Endocarditis (IE), Lyme Disease, Mycobacterial Infections, Otitis Media (OM), Pertussis, Streptococcal Pharyngitis, Tonsillitis streptococcal, Uncomplicated skin and subcutaneous tissue bacterial infections
Clarithromycin is a semi-synthetic, second-generation macrolide antibiotic belonging to the polyketide class of natural products.[1] Developed in 1980 and first approved for medical use in 1990, it is derived from erythromycin A through the methylation of the hydroxyl group at the C-6 position of the 14-membered lactone ring.[2] This specific structural modification was engineered to overcome key limitations of its parent compound, erythromycin. The primary advantages conferred by this change are enhanced stability in acidic environments, which significantly reduces the incidence of gastrointestinal side effects, and a more favorable pharmacokinetic profile, including improved oral absorption and a longer half-life.[5]
As a small molecule drug, clarithromycin has become a cornerstone in the treatment of a wide array of common bacterial infections.[1] It is widely prescribed for infections of the upper and lower respiratory tract, skin and soft tissues, and for the eradication of
Helicobacter pylori in peptic ulcer disease.[1] Its clinical utility is further extended to the management of opportunistic infections in immunocompromised individuals, most notably for the treatment and prophylaxis of disseminated
Mycobacterium avium complex (MAC) infection.[2]
Beyond its well-established antibacterial functions, clarithromycin exhibits significant immunomodulatory and anti-inflammatory properties that are independent of its antimicrobial activity.[5] These effects, which include the modulation of cytokine production, have positioned the drug as a subject of intensive research for potential applications in chronic inflammatory diseases and even as an adjunct in oncology.[10] More recently, an entirely distinct mechanism involving negative allosteric modulation of the GABA-A receptor has been identified, leading to its investigation as a novel therapy for central nervous system disorders of hypersomnolence.[7]
Despite its therapeutic versatility, the clinical use of clarithromycin is significantly constrained by two major factors. First, it is a potent inhibitor of the cytochrome P450 3A4 (CYP3A4) enzyme system, which results in a high potential for numerous and often severe drug-drug interactions.[6] Second, a notable cardiovascular safety concern has emerged, linking its use in patients with heart disease to an increased long-term risk of mortality.[7] These characteristics create a complex risk-benefit profile that requires careful clinical consideration.
Clarithromycin is chemically known as 6-O-methylerythromycin.[2] It presents as a white or almost white crystalline powder, though it can form colorless needles when crystallized from specific solvents like chloroform and diisopropyl ether.[2] The compound is practically insoluble in water but demonstrates solubility in organic solvents such as acetone and methylene chloride, and is slightly soluble in methanol, ethanol, and acetonitrile.[4] Its fundamental identifiers and properties are consolidated in Table 1 for reference.
Table 1: Key Identifiers and Physicochemical Properties of Clarithromycin
Identifier Type | Value | Source(s) |
---|---|---|
Generic Name | Clarithromycin | 1 |
English Name | Clarithromycin | 1 |
DrugBank ID | DB01211 | 1 |
CAS Number | 81103-11-9 | 2 |
Type | Small Molecule | 1 |
Molecular Formula | C38H69NO13 | 19 |
Molecular Weight (Average) | 747.95 g/mol | 2 |
Molecular Weight (Monoisotopic) | 747.47689126 Da | 2 |
PubChem CID | 84029 | 2 |
ChEMBL ID | CHEMBL1741 | 2 |
UNII | H1250JIK0A | 2 |
IUPAC Name | (3R,4S,5S,6R,7R,9R,11R,12R,13S,14R)-6-oxy-14-ethyl-12,13-dihydroxy-4-oxy-7-methoxy-3,5,7,9,11,13-hexamethyl-oxacyclotetradecane-2,10-dione | 2 |
Clarithromycin was developed in 1980 by the Japanese pharmaceutical company Taisho Pharmaceutical and was first approved for medical use in 1990.[6] Its development represents a significant milestone in the evolution of macrolide antibiotics.
The regulatory journey in the United States, overseen by the Food and Drug Administration (FDA), reflects a strategic expansion of its clinical applications. The original brand name, Biaxin, manufactured by Abbott Laboratories (now AbbVie), received its initial approval for the immediate-release tablet formulation on October 31, 1991.[22] This was followed by the approval of an oral suspension on December 23, 1993, to facilitate pediatric dosing.[22]
A key development in the drug's lifecycle was the approval of an extended-release formulation, Biaxin XL, on October 20, 2000.[23] This once-daily formulation was designed to improve patient adherence and convenience compared to the twice-daily regimen of the immediate-release version, a common strategy to maintain market relevance in the face of impending generic competition. Biaxin XL was initially approved for acute bacterial exacerbation of chronic bronchitis and acute maxillary sinusitis.[23] Its indications were further expanded on August 6, 2001, to include community-acquired pneumonia (CAP).[24] The approval pathway demonstrates a typical pharmaceutical product lifecycle, beginning with establishing a core market in common infections, expanding into more specialized and high-need indications like MAC and
H. pylori combination therapy, and finally innovating on the formulation to enhance patient convenience and extend the product's commercial viability.[22]
Today, clarithromycin is widely available as a generic medication from numerous manufacturers under various Abbreviated New Drug Applications (ANDAs).[25] Its global importance is underscored by its inclusion on the World Health Organization's List of Essential Medicines, which recognizes it as one of the safest and most effective medicines needed in a health system.[6]
The primary mechanism by which clarithromycin exerts its antibacterial effect is through the inhibition of protein synthesis in susceptible bacteria, a characteristic shared by all macrolide antibiotics.[1] The process begins with the drug's penetration of the bacterial cell wall. Once inside the cell, clarithromycin reversibly binds with high affinity to a specific site on the bacterial ribosome.[1]
The molecular target is Domain V of the 23S ribosomal RNA (rRNA), a critical structural and functional component of the 50S large ribosomal subunit.[1] In some organisms, such as
Shigella flexneri, the target has been more specifically identified as the Large ribosomal subunit protein uL10.[1] This binding event occurs within the polypeptide exit tunnel of the ribosome. By occupying this site, clarithromycin physically obstructs the passage of the nascent polypeptide chain. This leads to the inhibition of peptidyl transferase activity and blocks the translocation step of protein synthesis, where aminoacyl transfer-RNA (tRNA) moves from the A-site to the P-site of the ribosome.[1] The ultimate consequence is the premature dissociation of the incomplete peptide chain and a halt in the production of essential bacterial proteins, which prevents bacterial growth and replication.[1]
Clarithromycin is predominantly classified as a bacteriostatic agent, meaning it inhibits bacterial growth and multiplication rather than directly killing the organisms.[1] This allows the host's immune system to clear the contained infection. However, this classification is not absolute. Under certain conditions, clarithromycin can exhibit bactericidal (bacteria-killing) activity.[1] This dual activity is dependent on several factors, including the drug concentration at the site of infection and the specific susceptibility of the target organism.[1] At higher concentrations, the inhibition of protein synthesis can be profound enough to be lethal to the bacteria. Documented bactericidal activity has been observed against several clinically important pathogens, including
Haemophilus influenzae, Streptococcus pneumoniae, Streptococcus pyogenes, and Helicobacter pylori.[27]
Clarithromycin possesses a broad spectrum of antimicrobial activity that encompasses a wide range of clinically relevant pathogens. Its in-vitro potency is generally similar to or greater than that of its parent compound, erythromycin, against organisms that are susceptible to erythromycin.[1] The spectrum includes many Gram-positive and Gram-negative aerobic bacteria, as well as atypical organisms, some anaerobes, and mycobacteria. A detailed summary of its spectrum of activity is provided in Table 2.
Table 2: Spectrum of Antimicrobial Activity of Clarithromycin
Category | Organism | Clinical Relevance/Note | Source(s) |
---|---|---|---|
Gram-Positive Aerobes | Staphylococcus aureus | Active against methicillin-susceptible strains (MSSA) only; not active against MRSA. | 1 |
Streptococcus pneumoniae | Key pathogen in respiratory tract infections. | 1 | |
Streptococcus pyogenes (Group A) | Common cause of pharyngitis and skin infections. | 1 | |
Streptococcus agalactiae (Group B) | In-vitro activity demonstrated. | 7 | |
Listeria monocytogenes | In-vitro activity demonstrated. | 27 | |
Viridans group streptococci | In-vitro activity demonstrated. | 1 | |
Gram-Negative Aerobes | Haemophilus influenzae | Common respiratory pathogen. Efficacy enhanced by the active 14-OH metabolite. | 1 |
Haemophilus parainfluenzae | Respiratory pathogen. | 7 | |
Moraxella catarrhalis | Common cause of sinusitis, otitis media, and bronchitis exacerbations. | 1 | |
Helicobacter pylori | Primary target in peptic ulcer disease eradication therapy. | 1 | |
Legionella pneumophila | Causative agent of Legionnaires' disease. | 7 | |
Bordetella pertussis | Causative agent of whooping cough. | 7 | |
Neisseria gonorrhoeae | In-vitro activity demonstrated. | 27 | |
Campylobacter jejuni | In-vitro activity demonstrated. | 27 | |
Mycobacteria | Mycobacterium avium complex (MAC) | Critical indication for treatment and prophylaxis in immunocompromised patients. | 2 |
Mycobacterium leprae | Causative agent of leprosy. | 27 | |
Atypical Organisms | Mycoplasma pneumoniae | Common cause of atypical pneumonia. | 7 |
Chlamydia pneumoniae (TWAR) | Common cause of atypical pneumonia. | 1 | |
Chlamydia trachomatis | In-vitro activity demonstrated. | 27 | |
Ureaplasma urealyticum | In-vitro activity demonstrated. | 27 | |
Other Organisms | Toxoplasma gondii | Used in combination therapy for toxoplasmic encephalitis in HIV patients. | 1 |
Borrelia burgdorferi | Causative agent of Lyme disease; used as a second-line agent. | 1 | |
Bartonella species | Causative agent of cat scratch disease and other forms of bartonellosis. | 1 | |
Anaerobes | Clostridium perfringens | In-vitro activity demonstrated. | 7 |
Peptostreptococcus species | In-vitro activity demonstrated. | 27 | |
Cutibacterium acnes (formerly Propionibacterium acnes) | In-vitro activity demonstrated. | 7 |
Beyond its direct action on bacteria, clarithromycin possesses a range of non-antibiotic properties that are of increasing clinical and investigational interest. These effects suggest that the drug's therapeutic identity may be broader than that of a conventional anti-infective agent.
Clarithromycin is recognized for its significant immunomodulatory effects, a property characteristic of 14- and 15-membered macrolides but not 16-membered ones.[10] These actions are independent of its ability to inhibit bacterial protein synthesis and involve direct modulation of the host's immune response. The drug can suppress the production and gene expression of several key pro-inflammatory cytokines, including Interleukin-1β (IL-1β), IL-2, IL-5, IL-6, and Tumor Necrosis Factor-alpha (TNF-α).[5] For instance, by suppressing IL-1β, clarithromycin can indirectly reduce the production of IL-6, a critical growth factor for multiple myeloma cells.[10] Conversely, it has been shown to induce the production of the anti-inflammatory cytokine IL-4, which can inhibit IL-6 synthesis and reduce plasma cell growth.[10]
These anti-inflammatory actions form the basis for its successful use in chronic inflammatory pulmonary diseases such as diffuse panbronchiolitis and cystic fibrosis, where it can reduce neutrophil-driven inflammation and improve clinical outcomes.[29] This capacity to temper excessive host inflammatory responses has also led to its investigation as an adjunctive therapy in severe acute infections, including community-acquired pneumonia and COVID-19, with the goal of preventing progression to severe respiratory failure.[11] Furthermore, these pleiotropic effects are being explored in oncology, where clarithromycin is under investigation as an add-on therapy for hematologic malignancies like multiple myeloma, potentially through mechanisms such as autophagy inhibition and suppression of myeloma growth factors.[10]
A distinct and more recently discovered non-antibiotic mechanism of clarithromycin involves its interaction with the central nervous system. Emerging research has identified clarithromycin as a negative allosteric modulator of the gamma-aminobutyric acid type A (GABA-A) receptor.[7] GABA is the primary inhibitory neurotransmitter in the brain, and its action at the GABA-A receptor promotes sleep and sedation. By negatively modulating this receptor, clarithromycin is hypothesized to reduce GABAergic signaling, thereby producing a wakefulness-promoting (somnolytic) effect.[7]
This novel neurological mechanism is completely unrelated to its antibacterial function and has opened a new avenue of clinical investigation. Clarithromycin is being actively researched as a potential treatment for central disorders of hypersomnolence, such as idiopathic hypersomnia (IH) and narcolepsy type 2.[7] Clinical trials have demonstrated that clarithromycin can produce clinically meaningful improvements in subjective sleepiness in patients with these conditions, and the American Academy of Sleep Medicine has issued a conditional recommendation for its use in patients who are refractory to standard therapies.[7] This potential "repurposing" of an established antibiotic for a primary neurological disorder highlights a paradigm shift in understanding the drug's full therapeutic potential.
The clinical efficacy and safety of clarithromycin are governed by its pharmacokinetic properties—the processes of absorption, distribution, metabolism, and excretion (ADME). Its profile represents a significant improvement over its predecessor, erythromycin.
Following oral administration, clarithromycin is rapidly and effectively absorbed from the gastrointestinal tract.[1] A key chemical advantage is its stability in the presence of gastric acid, which allows for reliable oral absorption without the need for protective enteric coatings and contributes to its improved gastrointestinal tolerability compared to erythromycin.[2]
The absolute bioavailability of a 250 mg immediate-release clarithromycin tablet is approximately 50%, with the remaining fraction undergoing extensive first-pass metabolism.[4] The presence of food has a modest effect on the absorption of the immediate-release formulation. Food can slightly delay the onset of absorption, increasing the time to reach peak plasma concentration (Tmax) from approximately 2 hours to 2.5 hours. It may also increase the peak plasma concentration (Cmax) by about 24%, but it does not significantly alter the total extent of drug absorption, as measured by the area under the concentration-time curve (AUC).[4] In contrast, the administration instructions for the extended-release (XL) tablets specify that they should be taken with food to ensure proper absorption.[8]
Once absorbed into the systemic circulation, clarithromycin distributes readily into body tissues and fluids.[4] In plasma, it is approximately 70% bound to proteins.[1] A defining characteristic of clarithromycin, and macrolides in general, is its excellent tissue penetration, leading to concentrations in tissues that are substantially higher than those in the serum.[4]
The drug is actively transported into phagocytic cells, such as neutrophils and macrophages, which then migrate to sites of infection. During active phagocytosis, large concentrations of clarithromycin are released, effectively delivering the antibiotic directly to the location of the pathogens.[7] Tissue concentrations can be more than 10 times higher than concurrent plasma concentrations, with the highest levels observed in the liver, lung tissue (including epithelial lining fluid and alveolar macrophages), and stool.[5] This high degree of tissue penetration is fundamental to its efficacy in treating respiratory and soft-tissue infections.[39] While total drug concentrations in tissues are high, studies using in-vivo microdialysis have shown that the concentration of free, unbound drug in the interstitial fluid of subcutaneous and skeletal muscle tissue is lower than in plasma, with tissue-to-plasma free AUC ratios of approximately 0.4.[39]
Clarithromycin undergoes rapid and extensive first-pass metabolism in the liver following oral absorption.[7] The primary pathway for its metabolism is mediated by the cytochrome P450 3A4 (CYP3A4) isoenzyme, which is responsible for hydroxylation and N-demethylation reactions.[1] This metabolic pathway is of profound clinical importance because clarithromycin is not only a substrate of CYP3A4 but also a potent inhibitor of the enzyme.[1] This dual role as a substrate and inhibitor is the mechanistic basis for its numerous and clinically significant drug-drug interactions.
The principal metabolite formed is 14-(R)-hydroxyclarithromycin (14-OH clarithromycin).[1] This metabolite is not an inactive byproduct; it is microbiologically active and works synergistically with the parent compound to enhance its overall antibacterial effect.[1] The activity of the 14-OH metabolite varies depending on the target pathogen. For most organisms, it is less potent than the parent drug. However, it exhibits twofold greater activity than clarithromycin against
Haemophilus influenzae, a common respiratory pathogen.[5] This enhanced activity of the metabolite is a key contributor to clarithromycin's clinical efficacy in treating infections caused by this organism. Other, less significant metabolites, such as N-desmethylclarithromycin, are also formed.[7]
Clarithromycin and its metabolites are eliminated from the body through both renal and fecal routes.[4] For the immediate-release tablet formulation, approximately 20% to 30% of an administered dose is excreted unchanged in the urine. This fraction is higher, at around 40%, for the oral suspension formulation.[4] The active 14-OH metabolite accounts for an additional 10% to 15% of the dose recovered in the urine.[4] The remainder of the drug is eliminated through non-renal pathways, primarily in the feces after hepatic metabolism and biliary excretion.
The pharmacokinetics of clarithromycin are slightly non-linear, which is reflected in its dose-dependent elimination half-life.[4]
Steady-state plasma concentrations of both the parent drug and its active metabolite are typically achieved within 3 days of initiating a consistent dosing regimen.[4] A summary of these key pharmacokinetic parameters is presented in Table 3.
Table 3: Summary of Pharmacokinetic Parameters for Clarithromycin and its Active Metabolite
Parameter | Clarithromycin (Parent Drug) | 14-OH Clarithromycin (Active Metabolite) | Source(s) |
---|---|---|---|
Oral Bioavailability | ~50% | N/A | 7 |
Tmax (Immediate-Release) | ~2–2.5 hours | N/A | 4 |
Plasma Protein Binding | ~70% | N/A | 1 |
Half-life (t1/2) at 250 mg q12h | 3–4 hours | 5–6 hours | 4 |
Half-life (t1/2) at 500 mg q8-12h | 5–7 hours | 7–9 hours | 4 |
Primary Metabolism | Hepatic (CYP3A4) | N/A | 1 |
Primary Route of Elimination | Renal and Fecal | Renal | 4 |
The clinical use of clarithromycin is guided by its broad antimicrobial spectrum and favorable pharmacokinetic profile. It is indicated for a range of infections, with specific recommendations for dosing and duration varying by condition, patient population, and formulation. A guiding principle for its prescription is to reserve its use for infections that are proven or strongly suspected to be caused by susceptible bacteria, in order to mitigate the development of antimicrobial resistance.[38]
Clarithromycin is approved by the U.S. Food and Drug Administration (FDA) for the treatment of mild to moderate infections caused by designated susceptible microorganisms.[8]
This category represents a primary area of use for clarithromycin.
Clarithromycin is indicated for the treatment of uncomplicated skin and soft tissue infections caused by methicillin-susceptible Staphylococcus aureus or Streptococcus pyogenes.[1]
A critical indication for clarithromycin is its role in multi-drug regimens for the eradication of H. pylori, the bacterium responsible for most peptic ulcers.[1] To prevent the rapid development of resistance, it is always used in combination with other agents. Approved regimens include triple therapy with a proton pump inhibitor (such as omeprazole or lansoprazole) and another antibiotic (typically amoxicillin), or as part of a co-packaged triple therapy with vonoprazan and amoxicillin.[1]
Clarithromycin is a key agent in the management of infections caused by the Mycobacterium avium complex (MAC), particularly in immunocompromised individuals such as those with advanced HIV infection.[1] It is FDA-approved for both the treatment of active, disseminated MAC disease and for primary prophylaxis to prevent the initial occurrence of infection in high-risk patients (e.g., those with CD4 counts below 50 cells/mm³).[2]
In addition to its approved indications, clarithromycin is used for several other conditions based on clinical evidence and guideline recommendations.
Clarithromycin is available in several oral formulations to accommodate different patient needs and clinical scenarios.
Dosing for clarithromycin is highly dependent on the indication, patient age, severity of infection, and the formulation used. A summary of common FDA-approved dosing regimens is provided in Table 4.
Table 4: FDA-Approved Dosing Regimens for Clarithromycin by Indication
Indication | Patient Population | Formulation | Dose | Frequency | Duration | Source(s) |
---|---|---|---|---|---|---|
Community-Acquired Pneumonia | Adult | IR Tablet | 250 mg | Every 12 hours | 7–14 days | 8 |
Adult | XL Tablet | 1000 mg (2 x 500 mg) | Once daily | 7 days | 41 | |
Pediatric (≥6 mos) | Suspension | 7.5 mg/kg | Every 12 hours | 10 days | 8 | |
Acute Maxillary Sinusitis | Adult | IR Tablet | 500 mg | Every 12 hours | 14 days | 42 |
Adult | XL Tablet | 1000 mg (2 x 500 mg) | Once daily | 14 days | 41 | |
Acute Exacerbation of Chronic Bronchitis | Adult | IR Tablet | 250–500 mg | Every 12 hours | 7–14 days | 42 |
Adult | XL Tablet | 1000 mg (2 x 500 mg) | Once daily | 7 days | 41 | |
Pharyngitis / Tonsillitis | Adult | IR Tablet | 250 mg | Every 12 hours | 10 days | 42 |
Uncomplicated Skin Infection | Adult | IR Tablet | 250 mg | Every 12 hours | 7–14 days | 42 |
H. pylori Eradication | Adult | IR Tablet | 500 mg | Every 8–12 hours (regimen dependent) | 10–14 days | 8 |
MAC Prophylaxis / Treatment | Adult | IR Tablet | 500 mg | Twice daily | Ongoing | 8 |
Pediatric (≥20 mos) | Suspension | 7.5 mg/kg (up to 500 mg) | Twice daily | Ongoing | 8 | |
Acute Otitis Media | Pediatric (≥6 mos) | Suspension | 7.5 mg/kg | Every 12 hours | 10 days | 8 |
Clarithromycin clearance is reduced in patients with impaired renal function.
The clinical utility of clarithromycin is balanced by a well-defined safety profile that includes a range of adverse effects, significant drug-drug interactions, and a major cardiovascular warning. A thorough understanding of these risks is essential for its safe and effective use.
The cardiovascular effects of clarithromycin represent one of its most significant safety concerns.
Clarithromycin is known to prolong the QT interval on an electrocardiogram (ECG), a condition that affects cardiac repolarization.[7] This effect increases the risk of developing life-threatening ventricular arrhythmias, most notably torsades de pointes, which can lead to syncope and sudden cardiac death.[7] The risk is elevated in patients with underlying risk factors, including:
In February 2018, the U.S. FDA issued a pivotal drug safety communication that fundamentally altered the risk assessment for clarithromycin, especially in patients with cardiovascular disease.[7] This warning was based on a 10-year follow-up of the CLARICOR trial, a large, randomized, placebo-controlled study.[17]
Based on its safety profile, clarithromycin is contraindicated or requires significant caution in several patient populations.
Clarithromycin is involved in over 725 documented drug interactions, with 281 classified as major, meaning the risk of the interaction generally outweighs the benefit.[55]
The vast majority of clinically significant interactions are due to clarithromycin's potent inhibition of the hepatic microsomal enzyme CYP3A4 and the drug efflux transporter P-glycoprotein (P-gp).[1] By inhibiting these pathways, clarithromycin impairs the metabolism and clearance of numerous co-administered drugs that are substrates of CYP3A4 or P-gp. This leads to elevated plasma concentrations of the interacting drug, potentially resulting in toxicity.
A summary of the most critical drug interactions is provided in Table 5.
Table 5: Clinically Significant Drug-Drug Interactions with Clarithromycin
Interacting Drug Class | Specific Examples | Mechanism of Interaction | Clinical Implication / Consequence | Management Recommendation | Source(s) |
---|---|---|---|---|---|
HMG-CoA Reductase Inhibitors (Statins) | Simvastatin, Lovastatin, Atorvastatin | Potent inhibition of CYP3A4-mediated statin metabolism. | Markedly increased statin plasma concentrations, leading to a high risk of myopathy and potentially fatal rhabdomyolysis. | Contraindicated with simvastatin and lovastatin. Avoid or use with extreme caution and lowest possible dose for atorvastatin. Withhold statin during the course of clarithromycin therapy. Consider switching to a non-CYP3A4 metabolized statin (e.g., pravastatin, rosuvastatin). | 49 |
Oral Anticoagulants | Warfarin, Rivaroxaban, Apixaban, Dabigatran | Inhibition of CYP3A4 and/or P-gp decreases anticoagulant clearance. | Enhanced anticoagulant effect. For warfarin, this leads to an increased INR. For all, there is a significantly increased risk of major, potentially life-threatening hemorrhage. | Avoid combination if possible. If co-administration is necessary, perform close monitoring of INR (for warfarin) or for clinical signs of bleeding (for DOACs). Dose reduction of the anticoagulant may be required. | 14 |
Calcium Channel Blockers (CCBs) | Amlodipine, Nifedipine, Diltiazem, Verapamil | Inhibition of CYP3A4-mediated CCB metabolism. | Increased CCB plasma concentrations, leading to excessive vasodilation, hypotension, and a documented increased risk of hospitalization for acute kidney injury (secondary to renal hypoperfusion) and all-cause mortality, particularly in older adults. | Avoid combination whenever possible. If unavoidable, monitor blood pressure and renal function closely. Consider using an alternative, non-interacting antibiotic (e.g., azithromycin). | 48 |
QT-Prolonging Agents | Amiodarone, Quinidine, Sotalol, Pimozide, Disopyramide | Additive pharmacodynamic effect on cardiac repolarization (QT interval prolongation). | Substantially increased risk of life-threatening ventricular arrhythmias, including torsades de pointes. | Contraindicated with pimozide. Avoid concomitant use with other QT-prolonging drugs whenever possible. If combination is essential, ECG monitoring is warranted. | 14 |
Ergot Alkaloids | Ergotamine, Dihydroergotamine | Inhibition of CYP3A4-mediated metabolism. | Elevated ergot alkaloid levels, leading to acute ergot toxicity characterized by severe peripheral vasospasm, ischemia, and dysesthesia. | Contraindicated. | 14 |
Benzodiazepines | Triazolam, Midazolam (oral) | Inhibition of CYP3A4-mediated metabolism. | Increased plasma concentrations of the benzodiazepine, leading to prolonged and enhanced sedation and respiratory depression. | Monitor for excessive sedation. Dose adjustment of the benzodiazepine may be necessary. | 48 |
Theophylline | Theophylline | Inhibition of metabolism (mechanism less defined, may involve CYP1A2). | Increased serum theophylline levels, leading to a risk of theophylline toxicity (nausea, vomiting, seizures, arrhythmias). | Monitor serum theophylline concentrations and adjust dose as needed. | 14 |
Colchicine | Colchicine | Inhibition of CYP3A4 and P-gp decreases colchicine clearance. | Increased risk of severe, potentially fatal colchicine toxicity, especially in patients with renal or hepatic impairment. | Contraindicated in patients with renal or hepatic impairment. | 6 |
The efficacy of any antibiotic is fundamentally threatened by the ability of bacteria to develop resistance. For clarithromycin, this is a growing concern that has already reshaped clinical practice, particularly in the treatment of Helicobacter pylori.
Bacteria have evolved several mechanisms to evade the action of clarithromycin.
The rise of clarithromycin resistance has profound implications for its clinical use.
The increasing prevalence of clarithromycin resistance is now recognized as the single most important factor contributing to the failure of H. pylori eradication therapies.[66] The impact is dramatic: while standard clarithromycin-based triple therapy can achieve eradication rates of nearly 90% against susceptible strains, its efficacy plummets to as low as 18% against resistant strains.[69]
Resistance rates are not uniform and vary significantly by geographic region, but the global trend is one of a steady and alarming increase. In many parts of the world, including regions of Europe, Asia, and North America, the prevalence of primary clarithromycin resistance now exceeds the 15% threshold.[66] This threshold is a critical benchmark used in major clinical guidelines; once local resistance rates surpass this level, standard clarithromycin-based triple therapy is no longer recommended as a reliable first-line empiric treatment.[66] In recognition of this growing threat, the World Health Organization (WHO) has included clarithromycin-resistant
H. pylori on its priority list of pathogens for which new antibiotics are urgently needed.[66]
The failure of what was once a "gold standard" therapy has necessitated a complete overhaul of clinical practice guidelines for H. pylori infection. This evolution serves as a powerful case study in the real-world impact of antibiotic resistance.
Clarithromycin is not a naturally occurring compound; it is a semi-synthetic macrolide produced through the chemical modification of Erythromycin A, which is itself a product of bacterial fermentation.[5] The defining synthetic step is the selective methylation of the hydroxyl group at the 6-position of the erythromycin lactone ring, resulting in the chemical entity 6-O-methylerythromycin.[2]
This targeted methylation is structurally and functionally crucial. In the acidic environment of the stomach, the 6-OH group of erythromycin can participate in an intramolecular reaction with the C9-keto group to form an inactive 6,9-hemiketal and subsequently a 6,9;9,12-spiroketal. This degradation pathway is responsible for erythromycin's acid instability and is a major contributor to its gastrointestinal side effects. By methylating the 6-OH group, this internal reaction is blocked, rendering clarithromycin stable in gastric acid and thereby improving its oral bioavailability and tolerability.[5]
The primary challenge in the industrial synthesis of clarithromycin is achieving this methylation with high regioselectivity. The erythromycin molecule contains several other hydroxyl groups (e.g., at the C11, C12, and 4" positions) that are also susceptible to methylation. Non-selective methylation would lead to the formation of multiple undesired by-products, complicating purification and reducing the overall yield of the final product.[76]
To address the challenge of selective methylation, modern manufacturing processes employ a multi-step strategy of protection, methylation, and deprotection, as detailed in various patents.[75] A representative synthetic route includes the following key stages:
Various patents describe optimizations of this general pathway, including the use of different protecting groups, solvent systems, and "one-pot" procedures designed to improve efficiency, increase overall yield, and reduce the generation of impurities and environmental waste.[75]
The original developer and marketer of clarithromycin was Abbott Laboratories (which later spun off its pharmaceutical division into AbbVie), under the well-known brand name Biaxin.[22] Following the expiration of its patents, the market has opened to a vast number of generic manufacturers worldwide.
Major global producers of generic clarithromycin include large pharmaceutical companies such as AdvaCare Pharma, Apotex, Aurobindo Pharma, Mylan, Sandoz, and Teva Pharmaceuticals, as well as a multitude of manufacturers based in China, India, and Europe.[78]
Reflecting its widespread global use, clarithromycin is marketed under hundreds of different brand names across various countries. In addition to Biaxin, some of the most common international brand names include Klaricid, Klacid, and Xetinin XL.[1] The sheer number of available brands and suppliers underscores its status as a globally important and widely prescribed antibiotic.
Despite being on the market for over three decades, clarithromycin continues to be the subject of active clinical investigation, a testament to its complex pharmacology. Current research is largely focused on harnessing its non-antibacterial properties for novel therapeutic applications, moving far beyond its original role as an anti-infective.
Clarithromycin occupies a complex and evolving position in the therapeutic armamentarium. It was introduced as a pivotal second-generation macrolide that offered clear advantages in acid stability and tolerability over erythromycin, quickly establishing itself as a workhorse antibiotic for a multitude of common infections. For decades, it has been a first-line or key alternative agent for respiratory tract infections, skin infections, and, most notably, as part of the revolutionary multi-drug regimens that made the cure of H. pylori-induced peptic ulcer disease possible.
However, the clinical landscape for clarithromycin is now defined by two formidable challenges that are progressively constraining its traditional role. The first is the relentless global rise of antibiotic resistance, which has had its most dramatic impact on the treatment of H. pylori. The drug's efficacy has been so severely eroded that what was once a gold-standard therapy is now recommended against for empiric use in many regions, a stark illustration of the consequences of antimicrobial resistance. The second challenge is its intricate safety profile, which is dominated by its potent inhibition of CYP3A4—leading to a vast and hazardous web of drug-drug interactions—and a serious FDA warning regarding an unexplained increase in long-term cardiovascular mortality in patients with heart disease.
In this context, the future of clarithromycin may lie less in its utility as a frontline antibiotic and more in the strategic exploitation of its "secondary" non-antimicrobial properties. The ongoing, high-quality research into its immunomodulatory effects in sepsis and its neuro-modulatory actions in hypersomnia could potentially redefine its place in medicine. This evolution—from a straightforward anti-infective to a complex modulator of host biology—encapsulates the dynamic and often unpredictable lifecycle of a pharmaceutical agent. The story of clarithromycin thus serves a dual purpose: it is a potent reminder of the critical need for antibiotic stewardship to preserve the efficacy of our existing anti-infectives, while also demonstrating the remarkable potential for established molecules to find new life in treating conditions far beyond their original purpose.
Published at: July 21, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.