Meropenem is a synthetic, broad-spectrum carbapenemantibiotic indicated for the treatment of serious bacterial infections, including complicated skin and skin structure infections and bacterial meningitis in pediatric patients aged 3 months and older, as well as intra-abdominal infections (including in neonates under 3 months).[1] It exerts bactericidal activity by binding to penicillin-binding proteins, thereby inhibiting bacterial cell wall synthesis and leading to cell death; it is particularly stable against many β-lactamases produced by Gram-positive and Gram-negative bacteria.[2][1] Administered intravenously, meropenem achieves peak plasma concentrations of approximately 23 mcg/mL after a 500 mg dose and 49 mcg/mL after a 1 g dose over a 30-minute infusion, with a half-life of about 1 hour in adults with normal renal function, and is primarily excreted unchanged in the urine.[1]Developed by Sumitomo Pharmaceuticals as a more stable derivative of the natural carbapenem thienamycin (discovered in 1976 from Streptomyces cattleya), meropenem features a 1β-methyl group that confers resistance to renal dehydropeptidase I hydrolysis, eliminating the need for a co-administered inhibitor like cilastatin (used with imipenem).[3] First approved by the U.S. Food and Drug Administration in 1996 under the brand name Merrem IV, it is available as a sterile powder for reconstitution in 500 mg and 1 g vials containing meropenem trihydrate and sodium carbonate.[1] Unlike earlier carbapenems, meropenem demonstrates enhanced activity against certain Gram-negative pathogens, such as Pseudomonas aeruginosa, while maintaining efficacy against methicillin-susceptible staphylococci and streptococci.[2][3]Meropenem is reserved for infections caused by susceptible bacteria to minimize the risk of resistance development, and its use requires caution in patients with hypersensitivity to β-lactams, seizure disorders, or renal impairment due to potential adverse effects including anaphylaxis, convulsions, and thrombocytopenia.[1] In 2017, a fixed-dose combination of meropenem with vaborbactam (Vabomere) was approved for complicated urinary tract infections resistant to other antibiotics, highlighting its role in combating multidrug-resistant pathogens.[4]
Medical uses
Indications
Meropenem is a broad-spectrum carbapenem antibiotic with activity against aerobic and anaerobic Gram-positive and Gram-negative bacteria, including Pseudomonas aeruginosa.[5][6]It is indicated for the treatment of complicated skin and skin structure infections in adults and pediatric patients aged 3 months and older, caused by susceptible isolates such as methicillin-susceptible Staphylococcus aureus, Streptococcus pyogenes, and Escherichia coli.[1] Meropenem is also approved for complicated intra-abdominal infections in adults and children aged 3 months and older due to pathogens including E. coli, Klebsiella pneumoniae, P. aeruginosa, and Bacteroides fragilis.[1] Additionally, it is indicated for bacterial meningitis in pediatric patients aged 3 months and older caused by Haemophilus influenzae, Neisseria meningitidis, or penicillin-susceptible Streptococcus pneumoniae, with evidence of efficacy in eliminating concurrent bacteremia.[1]Meropenem is recommended for empirical therapy in hospitalized patients with severe pneumonia, sepsis, and polymicrobial infections, as well as for febrile neutropenia in neutropenic patients suspected of bacterial infection.[7][8] The Infectious Diseases Society of America (IDSA) guidelines recommend meropenem as a preferred carbapenem for treating severe infections outside the urinary tract caused by extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae, based on clinical trial data showing lower mortality compared to alternatives like piperacillin-tazobactam.[9][10]Clinical trials have demonstrated high efficacy for meropenem in complicated intra-abdominal infections, with clinical cure rates of 93-94% at test-of-cure in microbiologically evaluable populations.[11] Overall response rates exceed 80% in severe cases, supporting its role as monotherapy for polymicrobial intra-abdominal infections.[12][13]
Limitations and resistance considerations
Meropenem exhibits limited activity against certain Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA) and Enterococcus faecium, which are uniformly resistant due to intrinsic mechanisms such as altered penicillin-binding proteins and efflux pumps.[14][15] It also lacks efficacy against most fungi and viruses, as it targets bacterial cell wall synthesis and does not affect eukaryotic or viral replication pathways. Additionally, meropenem has poor activity against atypical bacteria like Mycoplasma species, which lack cell walls and are thus unaffected by beta-lactam antibiotics.[16]A major limitation arises from the increasing prevalence of carbapenem resistance, primarily driven by the production of carbapenemase enzymes such as Klebsiella pneumoniae carbapenemase (KPC) and New Delhi metallo-beta-lactamase (NDM) in pathogens including Klebsiella pneumoniae and Acinetobacter baumannii. These enzymes hydrolyze the beta-lactam ring of meropenem, rendering it ineffective, and are often plasmid-mediated, facilitating rapid dissemination among Gram-negative bacteria. Resistance in A. baumannii may also involve additional mechanisms like efflux pumps and porin loss, exacerbating treatment challenges in hospital settings.[17][18][19]Global surveillance data highlight the escalating threat of carbapenem resistance. According to the World Health Organization's 2025 report, antibiotic resistance in Gram-negative bacteria has risen in over 40% of monitored pathogen-antibiotic combinations between 2018 and 2023, with Escherichia coli showing 44.8% resistance to third-generation cephalosporins (a proxy for broader beta-lactam resistance trends) and K. pneumoniae at 55.2%. In the United States, the Centers for Disease Control and Prevention reported a more than 460% surge in infections caused by NDM-producing carbapenem-resistant Enterobacterales (CRE) from 2019 to 2023, particularly affecting pneumonia and bloodstream infections. Resistance rates for carbapenem-non-susceptible Gram-negatives can reach 20-50% in high-burden regions like parts of Asia and the Middle East.[20][21][22]To mitigate resistance, antimicrobial stewardship programs emphasize reserving meropenem for confirmed multidrug-resistant infections, such as those caused by extended-spectrum beta-lactamase (ESBL)-producing or CRE pathogens, rather than empirical use in mild cases. Guidelines from the Infectious Diseases Society of America (IDSA) recommend avoiding meropenem in community-acquired pneumonia unless resistant pathogens are identified through microbiological testing, favoring narrower-spectrum agents like beta-lactam/beta-lactamase inhibitor combinations for initial therapy. The American Thoracic Society similarly advises against routine carbapenem use in non-severe community-acquired pneumonia to preserve efficacy against hospital-acquired resistant strains.[9][23][24]For infections involving meropenem-resistant strains, alternatives such as ceftazidime-avibactam are recommended, particularly for KPC-producing CRE, as avibactam inhibits serine-based beta-lactamases while ceftazidime provides broad Gram-negative coverage. IDSA guidance prioritizes ceftazidime-avibactam over carbapenems for susceptible resistant isolates, with susceptibility rates exceeding 90% against many multidrug-resistant Gram-negatives in clinical studies. Other options like meropenem-vaborbactam may be considered for specific enzyme profiles, but selection should be guided by local susceptibility patterns.[9][25][26]
Administration and dosing
Routes and forms
Meropenem is administered exclusively by the intravenous (IV) route, either as a short infusion over 15 to 30 minutes or as a bolus injection over 3 to 5 minutes, to ensure effective delivery and minimize vein irritation.[1] Intramuscular administration is not recommended, as it is not approved and can cause significant injection site pain due to the absence of local anesthetics in the formulation.[1]The primary dosage form is a sterile powder for reconstitution supplied in single-dose vials containing 500 mg or 1 g of meropenem (as the trihydrate) blended with anhydrous sodium carbonate.[1] For hospital settings, pre-mixed ready-to-use solutions are available in DUPLEX containers, which combine 500 mg or 1 g of meropenem with 50 mL of 0.9% sodium chloride injection, facilitating quicker preparation for full adult doses.[27]Preparation involves reconstituting the vial powder with sterile water for injection (10 mL for 500 mg vial or 20 mL for 1 g vial) to yield a 50 mg/mL concentration, followed by shaking and allowing it to stand until clear.[1] For infusion, the reconstituted solution is further diluted in compatible fluids such as 0.9% sodium chloride to concentrations of 1 to 20 mg/mL; it is compatible with central venous catheters for extended administration.[1] Post-reconstitution bolus solutions (up to 50 mg/mL) remain stable for 3 hours at room temperature (up to 25°C) or 13 hours refrigerated (5°C), while infusion solutions in 0.9% sodium chloride are stable for 1 hour at room temperature or 15 hours refrigerated; solutions must not be frozen.[1]For pediatric and neonatal use, the same vial sizes are employed, with reconstitution and dilution adjusted according to weight-based dosing requirements to accommodate smaller volumes.[1]
Dosage regimens
Meropenem is administered intravenously as the standard route for systemic infections. The recommended adult dosage is 500 mg every 8 hours for complicated skin and skin structure infections (1 g every 8 hours if Pseudomonas aeruginosa is suspected or documented), 1 g every 8 hours for complicated intra-abdominal infections, with a maximum of 2 g every 8 hours specifically for bacterial meningitis.[1][28][29]For patients with renal impairment, dosage adjustments are necessary to prevent accumulation due to the drug's primary renal excretion. In adults with creatinine clearance (CrCl) of 26-50 mL/min, the standard dose is reduced to every 12 hours; for CrCl 10-25 mL/min, half the standard dose is given every 12 hours; and for CrCl less than 10 mL/min, half the standard dose is administered every 24 hours.[1][28]
Creatinine Clearance (mL/min)
Dosage Adjustment (Adults)
>50
Standard dose every 8 hours
26-50
Standard dose every 12 hours
10-25
½ standard dose every 12 hours
<10
½ standard dose every 24 hours
Pediatric dosing in children aged 3 months and older with normal renal function is weight-based: 10 mg/kg (maximum 500 mg) every 8 hours for skin infections (20 mg/kg, maximum 1 g, if Pseudomonas aeruginosa is suspected), 20 mg/kg (maximum 1 g) every 8 hours for intra-abdominal infections, increasing to 40 mg/kg (maximum 2 g) every 8 hours for meningitis.[1][29] For neonates and infants under 3 months with intra-abdominal infections, doses range from 20 mg/kg every 12 hours (for preterm infants less than 32 weeks gestational age and postnatal age under 2 weeks) to 30 mg/kg every 8 hours (for term infants 32 weeks or more gestational age and postnatal age 2 weeks or more).[1]The duration of meropenem therapy generally ranges from 7 to 14 days, tailored to the site and severity of the infection, with clinical response guiding completion.[30] In critically ill patients, extended infusions over 3 hours are recommended to optimize the time above the minimum inhibitory concentration (MIC), improving pharmacokinetic/pharmacodynamic target attainment compared to standard 30-minute infusions.[31]Therapeutic drug monitoring is advised in cases of obesity or augmented renal clearance to ensure adequate exposure, as standard dosing may result in subtherapeutic levels in these populations.[32][33]
Contraindications and precautions
Absolute contraindications
Meropenem is absolutely contraindicated in patients with known hypersensitivity to meropenem itself, any component of the formulation, or other drugs in the carbapenem class.[34] This includes individuals with a documented history of anaphylaxis or severe allergic reactions to beta-lactam antibiotics, such as penicillins or cephalosporins, owing to the structural similarities that can lead to cross-reactivity.[34] Although the overall risk of cross-reactivity between penicillins and carbapenems like meropenem is low—estimated at less than 1% in patients with confirmed penicillin allergy—the potential for life-threatening hypersensitivity reactions necessitates strict avoidance in such cases.[35] The European Medicines Agency aligns with this guidance, prohibiting use in patients hypersensitive to meropenem or related beta-lactams.[36] No black-box warnings are associated with meropenem regarding contraindications as of 2025.[34]
Use in special populations
Limited published data on meropenem use in pregnant women are insufficient to inform a drug-associated risk for major birth defects and miscarriage; it should be used during pregnancy only if the potential benefits justify the risks, such as in cases of maternal sepsis or severe infections.[34] Animal reproduction studies in rats and monkeys at doses up to 3.2 times the maximum recommended human dose showed no adverse effects on embryofetal development or fertility.[34]Meropenem is excreted into breast milk in low concentrations, with average levels around 480 mcg/L and an estimated infant dose of 0.13% of the maternal weight-adjusted dose; it is generally considered compatible with breastfeeding, though infants should be monitored for potential gastrointestinal effects like diarrhea.[37] No adverse effects, such as thrush or dermatitis, have been reported in breastfed infants exposed to meropenem via milk.[37]In elderly patients, meropenem dosing requires adjustment based on renal function due to age-related declines in creatinine clearance, with no overall differences in safety or efficacy otherwise observed; they may also face a heightened risk of Clostridium difficile-associated diarrhea.[34] Routine assessment of renal function is recommended prior to and during therapy in this population.[34]For pediatric patients, meropenem is approved and safe for use in those aged 3 months and older for indications like bacterial meningitis, intra-abdominal infections, and skin infections, with dosing typically at 20 mg/kg every 8 hours; safety is not established in infants under 3 months except for specific intra-abdominal cases, and caution is advised in preterm neonates due to immature renal function.[34] No data exist on its use in pediatric patients with renal impairment.[34]In patients with renal impairment, meropenem doses must be reduced based on creatinine clearance (CrCl): for CrCl 26-50 mL/min, administer 1 g every 12 hours; for CrCl 10-25 mL/min, 500 mg every 12 hours; and for CrCl <10 mL/min, 500 mg every 24 hours, to avoid accumulation and toxicity like thrombocytopenia.[34] No dose adjustment is necessary for hepatic impairment, as pharmacokinetics remain unaffected.[34]In obese patients, particularly critically ill individuals, meropenem's volume of distribution increases with body mass index, but clearance is primarily driven by renal function; weight-based dosing using total body weight in CrCl calculations or extended infusions (e.g., 2 g every 8 hours over 3 hours) may be required to achieve adequate pharmacokinetic/pharmacodynamic targets against pathogens with higher minimum inhibitory concentrations.[38] Therapeutic drug monitoring is suggested for optimization in this group to ensure exposure.[38]
Adverse effects
Common side effects
Meropenem is generally well tolerated, with common side effects primarily involving the gastrointestinal tract, injection site, and mild laboratory abnormalities occurring in more than 1% of patients in clinical trials. Gastrointestinal disturbances are the most frequent, including diarrhea (reported in 2.5-4.8% of patients across studies involving over 5,000 individuals), nausea (3-5%), and vomiting (part of the 1.2-3.6% combined nausea/vomiting incidence). These effects are typically mild and transient, stemming from data in pivotal trials such as those reviewed by the Meropenem Serious Infection Study Group.[39][40][41]Injection site reactions, such as phlebitis, pain, inflammation, or rash, affect 1-3% of patients receiving intravenous administration, often related to the infusion process and resolving without intervention.[41][40]Hematologic and hepatic changes include mild elevations in liver enzymes, with increased ALT in 5-7% and AST in approximately 6% of adult patients from clinical trial data (n=2,904), alongside less frequent bilirubin elevations (around 2%). These are usually asymptomatic and reversible upon discontinuation.[41][42]Other common adverse effects encompass headache (2.3%) and non-severe rash (1.4-2%), both observed in multiple trials, as well as oral moniliasis in select pediatric populations. Management of these side effects is generally symptomatic; for instance, antiemetics can alleviate nausea and vomiting, while monitoring suffices for laboratory changes.[39][40][41]
Serious adverse effects
Meropenem, like other beta-lactam antibiotics, can rarely cause serious hypersensitivity reactions, including anaphylaxis and severe cutaneous adverse reactions such as Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). These reactions occur in less than 0.1% of patients and are more likely in individuals with a history of multiple drug allergies or prior beta-lactam hypersensitivity. The mechanism involves immune-mediated responses, potentially triggered by the beta-lactam ring structure, leading to rapid onset of symptoms like hypotension, bronchospasm, or widespread skin detachment; immediate discontinuation is required, and supportive care including epinephrine may be necessary.[43][44]Seizures represent another serious adverse effect of meropenem, with an incidence of approximately 0.5-1% in clinical trials, though rates can be higher (up to several percent) in patients with renal impairment, central nervous system disorders, or a history of epilepsy. This neurotoxicity is linked to meropenem's ability to penetrate the blood-brain barrier and potentially inhibit GABA-mediated inhibition in the central nervous system, exacerbating seizure risk in predisposed individuals. Monitoring for symptoms such as confusion or myoclonus is essential, particularly in high-risk groups, and dose adjustments based on renal function can mitigate this risk.[43][45]Clostridioides difficile-associated diarrhea (CDAD), ranging from mild to life-threatening colitis, can occur in meropenem-treated patients due to disruption of normal gut flora and overgrowth of toxin-producing C. difficile. This risk is inherent to broad-spectrum antibiotics like meropenem, which suppress beneficial bacteria, allowing pathogenic proliferation; prompt evaluation of persistent diarrhea is critical to prevent complications like pseudomembranous colitis. Incidence may vary by patient factors such as prolonged hospitalization or prior antibiotic exposure.[43][46]Hematologic abnormalities, including thrombocytopenia and neutropenia, are rare in clinical trials and are typically reversible upon discontinuation of meropenem. Thrombocytopenia is more common in patients with renal impairment, where reduced clearance may lead to drug accumulation and bone marrow suppression, while neutropenia can manifest as agranulocytosis in prolonged therapy. These effects stem from immune-mediated destruction or direct myelotoxicity, necessitating periodic blood count monitoring in at-risk patients.[43][40]Post-marketing surveillance has identified additional rare serious effects, including encephalopathy (manifesting as altered mental status or convulsions) and hemolytic anemia, often in patients with underlying conditions like renal failure or prolonged exposure. As of May 2025, rhabdomyolysis has also been reported, characterized by muscle pain, weakness, dark urine, or elevated creatine phosphokinase levels; discontinuation is recommended if suspected. These reports highlight the importance of vigilance beyond clinical trial data, with risk factors including advanced age, comorbidities, and concurrent medications that impair clearance. Adverse events should be reported to systems like FDA MedWatch to support ongoing pharmacovigilance and updates to safety profiles.[43][34]
Interactions
Drug-drug interactions
Meropenem exhibits several notable drug-drug interactions, primarily involving alterations in renal excretion, anticonvulsant levels, and synergistic antimicrobial effects. Co-administration with probenecid inhibits the renal tubular secretion of meropenem, leading to increased plasma concentrations by approximately 56% and prolongation of its half-life by 38%; this interaction necessitates avoidance or dose reduction of meropenem to prevent potential toxicity.[1]A significant pharmacokinetic interaction occurs with valproic acid, where meropenem reduces serum valproate concentrations by approximately 50-70%, potentially within 24 hours, through an unclear mechanism possibly involving enhanced metabolism or reduced absorption; this can precipitate breakthrough seizures, and concurrent use is generally not recommended, with close monitoring of valproate levels required if unavoidable.[47][48][49]Pharmacodynamically, meropenem demonstrates additive or synergistic activity when combined with aminoglycosides, such as gentamicin or tobramycin, particularly against Pseudomonas aeruginosa isolates, enhancing bactericidal effects against Gram-negative bacteria without notable pharmacokinetic interference.[50]Meropenem does not significantly interact with cytochrome P450 enzymes, minimizing risks with drugs metabolized via these pathways. Food has minimal effects on meropenem's absorption or efficacy, as it is administered intravenously. Clinical guidelines, such as those from prescribing information, recommend evaluating these interactions on a case-by-case basis and considering alternatives when possible.[51][5][1]
Other interactions
Meropenem can interfere with certain laboratory tests, potentially leading to inaccurate results that may affect clinical decision-making. It may cause false-positive results for urine glucose when using copper-reduction methods, such as Clinitest, due to its reducing properties as a beta-lactam antibiotic; enzymatic glucose oxidase methods, like Clinistix or Tes-Tape, are recommended instead to avoid this interference.[52] Additionally, meropenem has been associated with positive direct or indirect Coombs tests in post-marketing reports, which may indicate immune-mediated hemolysis and requires confirmation with alternative testing if hemolytic anemia is suspected.[1]No clinically significant interaction occurs with alcohol, allowing concurrent use without adjustment. Meropenem is compatible with dialysis, as it is hemodialyzable (removing about 30-50% per session), necessitating supplemental dosing—typically 500 mg—immediately after hemodialysis to maintain therapeutic levels.[53][54]Guidelines for laboratory monitoring during meropenem therapy include baseline and periodic evaluations of renal function (e.g., serum creatinine, eGFR), complete blood count (for thrombocytopenia or anemia), liver enzymes (for potential hepatotoxicity), and therapeutic drug monitoring of meropenem trough levels (targeting 2-4 mg/L) in critically ill patients or those with unstable pharmacokinetics to optimize efficacy and minimize toxicity.[1][55]
Pharmacology
Mechanism of action
Meropenem, a carbapenem-class β-lactam antibiotic, exerts its bactericidal action by binding covalently to penicillin-binding proteins (PBPs) in susceptible bacteria, thereby inhibiting the final transpeptidation step in peptidoglycan cross-linking during cell wall synthesis. This disruption weakens the bacterial cell wall, activating endogenous autolysins that cause cell lysis and death.[56][57]Meropenem demonstrates high affinity for key PBPs in both Gram-positive and Gram-negative bacteria, including PBPs 2, 3, and 4 in Escherichia coli and Pseudomonas aeruginosa, as well as PBPs 1, 2, and 4 in Staphylococcus aureus. However, it exhibits low affinity for PBP2a, the mecA-encoded PBP responsible for methicillin resistance in MRSA, which accounts for its limited activity against this pathogen.[56][58]The antibiotic's structural features confer stability against hydrolysis by many β-lactamases, such as AmpC cephalosporinases produced by Enterobacteriaceae and extended-spectrum β-lactamases (ESBLs) like CTX-M variants, allowing effective penetration and target engagement in resistant strains. In contrast, meropenem is susceptible to hydrolysis by carbapenemases, including class A enzymes like KPC and class B metallo-β-lactamases like NDM, which rapidly degrade it and confer resistance.[59][60]For susceptible Gram-negative bacteria such as E. coli, meropenem achieves low minimum inhibitory concentrations (MICs), typically ≤0.25 mg/L, reflecting its potent inhibition at sub-micromolar levels.[61]
Pharmacodynamics
Meropenem, as a carbapenem beta-lactam antibiotic, demonstrates time-dependent antibacterial activity, where its efficacy is primarily determined by the duration of exposure rather than peak concentrations. The key pharmacodynamic index is the percentage of the dosing interval during which the unbound (free) drug concentration exceeds the minimum inhibitory concentration (fT>MIC), with targets of greater than 40% fT>MIC required for bacteriostatic effects and 40-50% fT>MIC for bactericidal activity against susceptible pathogens.[62] Studies in animal models and in vitro simulations have shown that achieving 100% fT>MIC correlates with 90% maximal bactericidal efficacy, particularly for Gram-negative bacteria, underscoring the benefit of dosing strategies like extended infusions to prolong this exposure.[63]Meropenem also exhibits a post-antibiotic effect (PAE), a persistent suppression of bacterial regrowth following short-term drug exposure, which extends its therapeutic window. Against Gram-negative bacteria such as Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa, the PAE duration is concentration-dependent, typically ranging from 1 to 2 hours at concentrations around 4-8 times the MIC, though it can extend to 2-5 hours in some strains.[64] This effect is shorter against Gram-positive organisms but contributes to less frequent dosing needs compared to other beta-lactams without notable PAE.[65]The inoculum effect represents a limitation in meropenem's pharmacodynamics, where activity diminishes at high bacterial densities due to increased MICs and reduced drug efficacy. In vitro models simulating high inoculum sizes (e.g., >10^7 CFU/mL) show markedly reduced killing rates and potential for resistance emergence against pathogens like Klebsiella pneumoniae, as the drug's beta-lactamase stability is overwhelmed by greater beta-lactamase production or altered target expression.[66] This phenomenon highlights the need for combination therapy in infections with heavy bacterial loads, such as abscesses or biofilms.Synergistic interactions enhance meropenem's pharmacodynamics against challenging bacterial states. Combinations with aminoglycosides like tobramycin or fluoroquinolones like ciprofloxacin demonstrate additive or synergistic effects, improving penetration and killing within biofilms formed by Pseudomonas aeruginosa or Acinetobacter baumannii, where monotherapy fails due to matrix barriers.[67] Similarly, meropenem paired with agents like sulbactam shows enhanced activity against intracellular pathogens, such as Acinetobacter species in phagocytes, by overcoming limited drug penetration into host cells.[68] These synergies are supported by PK/PD models indicating improved fT>MIC attainment and reduced regrowth in co-culture systems.[69]
Pharmacokinetics
Meropenem is administered intravenously due to its poor oral bioavailability, resulting in negligible absorption through the gastrointestinal tract, while intravenous administration achieves 100% bioavailability.[1]Following intravenous infusion, meropenem exhibits a volume of distribution of approximately 0.25 L/kg in adults, indicating primarily extracellular distribution. It demonstrates good penetration into various tissues, including the cerebrospinal fluid (up to 30-40% of plasma concentrations in the presence of inflamed meninges), lungs, and soft tissues. Plasma protein binding is low at about 2%, which facilitates its availability for antibacterial activity at infection sites.[70][71][1]Meropenem undergoes minimal metabolism and is not metabolized by hepatic cytochrome P450 enzymes; approximately 70% (range 50-75%) of the dose is excreted unchanged in the urine, with the remainder recovered as an inactive open-β-lactam metabolite formed through chemical hydrolysis and minor enzymatic pathways.[16][1]Elimination is predominantly renal, with approximately 70% of the dose excreted unchanged in the urine via glomerular filtration and tubularsecretion. The elimination half-life is about 1 hour in adults with normal renal function but is prolonged in patients with renal impairment, necessitating dosage adjustments to maintain therapeutic concentrations relative to the time above minimum inhibitory concentration (fT>MIC).[1]
Chemistry
Chemical structure and properties
Meropenem has the molecular formula C₁₇H₂₅N₃O₅S and a molecular weight of 383.5 g/mol.[2] Its IUPAC name is (4R,5S,6S)-3-[(3S,5S)-5-(dimethylcarbamoyl)pyrrolidin-3-yl]sulfanyl-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid, and the canonical SMILES notation is C[C@@H]1[C@@H]2C@HC@@HO.[2]The chemical structure of meropenem features a bicyclic β-lactam ring fused to a pyrroline ring, characteristic of carbapenem antibiotics, with key side chains including a thio-linked pyrrolidine ring bearing a dimethylcarbamoyl group at the 5-position and a 1-hydroxyethyl substituent at the 6-position.[2] This β-lactam core provides the structural basis for its antibacterial activity by mimicking the D-alanyl-D-alanine substrate in bacterial cell wall synthesis.[5]Meropenem appears as a white to pale yellow crystalline powder.[2] It exhibits pKa values of approximately 2.9 for the carboxylic acid group and 7.4 for the pyrrolidine group.[72]In terms of solubility, meropenem is sparingly soluble in water (approximately 5.63 g/L), very slightly soluble in hydrated ethanol, and practically insoluble in acetone or ether.[2] The compound demonstrates greater stability in acidic conditions (pH around 6–6.5) compared to alkaline environments, where β-lactam hydrolysis accelerates degradation.[73]
Synthesis and stability
Meropenem is synthesized through a multi-step semi-synthetic process starting from thienamycin, a natural carbapenem antibiotic produced via fermentation of Streptomyces cattleya. The process involves protection of the β-lactam ring to prevent degradation during subsequent reactions, followed by attachment of the protected (3S,5S)-5-(dimethylcarbamoyl)pyrrolidin-3-yl thiol side chain via nucleophilic thioether formation, and final deprotection steps to yield the active trihydrate form.[74][75][76]The original synthesis was developed by Sumitomo Pharmaceuticals, with key patents such as US 4,943,569 covering the core process, which expired in 2010, enabling generic production. Generic routes typically employ similar chemical strategies, often optimized for higher yields and avoiding cryogenic conditions, while retaining the thienamycin fermentation precursor step.[77][78]Meropenem exhibits chemical instability primarily due to hydrolysis of the β-lactam ring amide bond, analogous to enzymatic β-lactamase mechanisms but occurring non-enzymatically in aqueous environments. As a sterile powder for injection, it maintains stability with a shelf life of 2–3 years when stored at controlled room temperature (15–30°C), protected from light and moisture.[79][80][81]In solution after reconstitution, stability is markedly reduced. For IV infusion administration (1–20 mg/mL) in 0.9% sodium chloride, solutions are stable for up to 1 hour at room temperature (≤25°C) or 15 hours refrigerated (≤5°C); in 5% dextrose, use immediately. For IV bolus administration (50 mg/mL), up to 3 hours at room temperature or 13 hours refrigerated. Do not freeze. Beyond these times, potency may drop below 90%. Major degradation products include open-ring penems formed by β-lactam hydrolysis and dimeric species from intermolecular aminolysis, which are microbiologically inactive and potentially immunogenic. Storage recommendations specify refrigeration at 2–8°C for reconstituted solutions to minimize degradation, with immediate use preferred after preparation.[1][72][82][83]Per the United States Pharmacopeia (USP) monograph, meropenem for injection must meet strict impurity limits, including not more than 0.8% for the primary degradant at relative retention time 0.45 and 0.6% for another at relative retention time 1.9, ensuring product purity through chromatographic analysis. Total unspecified impurities are limited to 1.0%, with overall related substances not exceeding 3.0% to maintain therapeutic safety and efficacy.[84][85]
History
Development and discovery
Meropenem's development originated from the discovery of thienamycin, a naturally occurring carbapenem antibiotic isolated in 1976 from the soil bacterium Streptomyces cattleya by researchers at Merck & Co. Inc. during a screening program for novel beta-lactam compounds.[86] Thienamycin exhibited potent broad-spectrum antibacterial activity but suffered from rapid degradation by renal dehydropeptidase I, limiting its clinical utility and prompting efforts to create more stable synthetic derivatives.[87]In response, pharmaceutical companies pursued structural modifications to enhance stability and reduce toxicity. Imipenem, a semi-synthetic analog of thienamycin developed by Merck, addressed some instability but still required co-administration with the dehydropeptidase inhibitor cilastatin to prevent renal metabolism and associated nephrotoxicity.[88] Sumitomo Pharmaceuticals, seeking a carbapenem that could be used without such a companion drug, initiated research in the early 1980s to design compounds resistant to enzymatic degradation while maintaining efficacy. The team, led by chemists including Makoto Sunagawa, focused on introducing a 1β-methyl group at the carbapenem core— an innovation initially explored by Lederle Laboratories— to sterically hinder dehydropeptidase access, combined with a (2S,4S)-2-[(3S)-3-carboxy-1-pyrrolidinylcarbonyl]pyrrolidin-4-ylthio side chain at the C-2 position to minimize neurotoxic potential.[89] Akira Yoshida and colleagues at Sumitomo contributed key synthetic methodologies, such as side-chain substitution reactions on 2-arylsulfinyl intermediates, enabling efficient production of these modified structures.[90]The first synthesis of meropenem (initially coded as SM-7338) was achieved in the late 1980s through these efforts, with a pivotal patent (US 4,943,569) filed by Sumitomo on October 8, 1987, describing the compound and its preparation.[91] Preclinical evaluations in animal models, including mice, rats, and dogs, demonstrated meropenem's superior stability and broad-spectrum activity against Gram-positive and Gram-negative pathogens compared to imipenem, with notably lower seizure induction in neurotoxicity assays—attributed to the optimized side chain reducing GABA antagonism.[92] Early efficacy data from systemic infection models, such as those against Pseudomonas aeruginosa and Staphylococcus aureus, confirmed protective doses comparable or better than imipenem without the need for enzyme inhibition, supporting its advancement. These findings, detailed in structure-activity relationship studies published in 1990, underscored meropenem's potential as a standalone injectable antibiotic.[89]
Regulatory approvals and milestones
Meropenem underwent phase III clinical trials in the 1990s, including randomized, double-blind studies comparing its efficacy to imipenem/cilastatin for treating intra-abdominal infections, where it demonstrated non-inferiority in clinical response rates.[93] These trials involved hospitalized patients with serious bacterial infections and supported meropenem's broad-spectrum activity against Gram-positive, Gram-negative, and anaerobic pathogens.[94]The European Medicines Agency granted initial marketing authorization for meropenem (as Meronem) on July 12, 1997, for indications including complicated intra-abdominal and skin infections in adults.[95]In the United States, the Food and Drug Administration approved meropenem (as Merrem IV) on June 21, 1996, initially for complicated skin and skin structure infections, intra-abdominal infections, and meningitis in adults and children aged 3 months and older.[79] Expanded pediatric indications followed, including approval for abdominal infections in infants under 3 months in 2015.[96]Key milestones include meropenem's inclusion on the World Health Organization's Model List of Essential Medicines in 2017, recognizing its role in treating severe infections like neutropenia in cancer patients.[97] The original U.S. patent expired around 2010, leading to the first generic approval by the FDA that year, which increased accessibility and reduced costs.[98] Pediatric exclusivity extensions have been granted under the Best Pharmaceuticals for Children Act, providing six-month market protections to incentivize studies in pediatric populations.[99]Post-approval developments emphasize antimicrobial stewardship, with meropenem subject to restrictions in many hospital programs to curb carbapenem-resistant infections, such as requiring infectious disease consultation for initiation.[100] No major withdrawals or bans have occurred, though ongoing monitoring addresses emerging resistance patterns through updated susceptibility testing guidelines.[101]
Society and culture
Trade names and branding
Meropenem is primarily marketed under the brand name Merrem in the United States and Meronem internationally, with these trademarks originally held by AstraZeneca following a licensing agreement with Sumitomo Pharma in the 1990s.[1][36][102] In 2016, Pfizer acquired the global commercialization rights to Merrem and Meronem (excluding certain Asian markets) from AstraZeneca, continuing its distribution as a broad-spectrum carbapenem antibiotic for severe hospital-treated infections.[103]Regionally, meropenem is sold under names such as Meropen in Japan by Sumitomo Pharma and various generics in India, including Meronem (formerly AstraZeneca) and Merolan (Mylan).[102][104] Worldwide, over 50 trade names exist for meropenem products, reflecting its availability through multiple manufacturers in diverse markets.[105]Upon patent expiration in the early 2010s, generic versions proliferated, produced by companies including Teva Pharmaceuticals, Mylan (now part of Viatris), and Sandoz (a Novartis division), enabling broader access to non-branded formulations.[106][107] The drug was branded and launched as a next-generation carbapenem, emphasizing its stability against renal dehydropeptidase-I and broad coverage against Gram-positive, Gram-negative, and anaerobicbacteria compared to predecessors like imipenem.[108] Packaging is designed for institutional use, typically as sterile powder vials for intravenous reconstitution in hospital settings, with no direct-to-consumer marketing.[1]
Availability and legal status
Meropenem is available exclusively by prescription in major markets, including the United States (Rx-only), Australia (S4), Canada (Rx-only), and the United Kingdom (POM), where it is administered intravenously in hospital settings due to its injection-only formulation. Globally, it is widely stocked in hospitals for treating severe infections, but retail availability is limited, particularly in low- and middle-income countries (LMICs), where public sectoraccess averages around 40% for essential antibiotics like meropenem. In LMICs, supply constraints and infrastructure challenges further restrict over-the-counter or community-level distribution, confining use primarily to tertiary care facilities.As an antibiotic, meropenem is not subject to controlled substance scheduling akin to narcotics; however, its use is regulated through antimicrobial stewardship programs (ASPs) worldwide to mitigate resistance development. These programs, implemented in hospitals and health systems, involve prospective audits, dosing optimization, and restriction protocols to ensure appropriate prescribing, with studies showing reductions in meropenem consumption by up to 50% without compromising outcomes. In the European Union and other regions, ASPs are mandated under broader antibiotic resistance action plans.In the United States as of 2025, generic meropenem pricing ranges from approximately $5 to $20 per gram for 1 g vials, while branded versions like Merrem can cost $50 to $100 per gram depending on supplier and volume. The World Health Organization (WHO) prequalified its first meropenem product in 2020, facilitating affordable generic access in LMICs through vetted manufacturers, which has helped lower costs to under $10 per gram in supported programs. Meropenem is included on the WHO Model List of Essential Medicines, promoting its procurement for critical conditions like sepsis.Supply shortages of meropenem occurred periodically between 2020 and 2022, primarily due to active pharmaceutical ingredient (API) disruptions from manufacturing delays and global supply chain issues exacerbated by the COVID-19 pandemic. These shortages led to increased use of alternatives and heightened costs in affected regions, with distribution often coordinated through UN agencies like UNICEF to stabilize supplies in LMICs. Access initiatives, such as WHO's sepsis management guidelines and essential medicines procurement mechanisms, support its availability in developing regions for treating multidrug-resistant infections, though gaps persist in rural and low-resource settings.
Research directions
Emerging indications
Meropenem is being investigated for several investigational applications beyond its established uses, particularly in complex infections where bacterial co-pathologies or resistance complicate treatment. Recent clinical trials from 2020 onward have explored its role in managing bacterial complications associated with viral pandemics and multidrug-resistant conditions, leveraging its broad-spectrum activity against Gram-negative pathogens.In the context of COVID-19, meropenem has shown potential as an adjunct for treating ventilator-associated pneumonia (VAP) in patients with bacterial co-infections, with studies between 2020 and 2023 highlighting its utility in critically ill cohorts despite challenges like resistance emergence. For instance, case reports and observational data indicate challenges with meropenem-resistant pathogens in secondary bacterial pneumonias in intubated COVID-19 patients, with outcomes varying with pathogen susceptibility.[109][110]Combination therapy with meropenem and rifampin has demonstrated promising early bactericidal activity against multidrug-resistant tuberculosis (MDR-TB) in phase II studies. The COMRADE trial, a randomized phase 2A study, evaluated meropenem (with clavulanate) at doses up to 6 g daily, alone or with rifampin, in patients with drug-susceptible pulmonary TB, reporting a median daily fall in colony-forming units of 0.22 log10 CFU/mL for the 2 g three-times-daily combination arm, indicating significant activity comparable to standard regimens but limited by gastrointestinal tolerability issues. These findings support further exploration of beta-lactam-based regimens for shortening MDR-TB treatment durations.[111]Exploratory applications of meropenem target biofilm-related infections, such as those in prosthetic joint infections (PJI), where its penetration into synovial fluid enhances eradication of adherent pathogens. A 2024 study on single-stage revision surgery combined with intra-articular meropenem infusion (50,000 µg/mL) achieved mean peak synovial concentrations of 5,819.1 µg/mL, exceeding the minimum biofilm eradication concentration and sustaining therapeutic levels for up to 8 days without significant adverse events. Additionally, case reports describe successful adjunct use with personalized phage therapy against Pseudomonas aeruginosa biofilms in chronic PJI, achieving infection resolution when standard antibiotics failed.[112][113]Limited data exist on meropenem's synergies in bacterial superinfections complicating viral diseases, positioning it as a potential adjunct to mitigate secondary bacterial invasions in conditions like COVID-19pneumonia. Reviews of therapeutic options note its role in addressing carbapenem-resistant superinfections amid viralinflammation, though clinical evidence remains preliminary and focused on empirical use in severe cases.[114]Ongoing trials as of 2024-2025, such as the BALANCE+ platform study (NCT05893147), are evaluating treatment strategies for Gram-negative bloodstream infections, including potential extensions to endocarditis, to optimize outcomes in high-risk populations where antibiotics like meropenem may be used. A recent case of Pseudomonas aeruginosa prosthetic valve endocarditis further illustrates meropenem's efficacy in combination with ceftolozane/tazobactam, achieving rapid bacteremia clearance and sterile valve cultures post-treatment.[115][116]
Resistance and novel formulations
Meropenem resistance, primarily driven by carbapenemase-producing Enterobacterales (CPE), has prompted the development of beta-lactamase inhibitor combinations to restore its efficacy. Meropenem-vaborbactam, approved by the FDA in 2017 for complicated urinary tract infections and intra-abdominal infections caused by susceptible strains, targets class A carbapenemases like KPC through vaborbactam's inhibition of beta-lactamase enzymes.[117] Recent expansions include ongoing trials evaluating its use in pediatric populations and against carbapenem-resistant Enterobacterales (CRE), demonstrating promising safety and microbiological cure rates in severe KPC-producing infections as of 2025.[118] Similarly, imipenem-relebactam, though not directly paired with meropenem, represents a parallel approach with relebactam inhibiting class A and C beta-lactamases, informing broader combo strategies against meropenem-resistant pathogens.[119]Novel formulations aim to enhance meropenem's delivery, particularly for lung infections where systemic administration limits local concentrations. Liposomal encapsulation of meropenem has shown improved stability and antibacterial activity against multidrug-resistant bacteria, with formulations combining it with biosurfactants like rhamnolipids achieving sustained release and reduced dosing frequency in preclinical models as of 2025.[120] Inhaled antibiotic strategies for ventilator-associated pneumonia often incorporate meropenem alongside agents like colistin.[121] These approaches build on stability insights from meropenem's chemical properties to overcome degradation barriers in respiratory environments.[122]Genomic surveillance of carbapenemase spread utilizes advanced tools like CRISPR-based systems to track resistance genes in real-time. Studies employing CRISPR-Cas dynamics reveal that isolates lacking these systems are more prone to acquiring mobile genetic elements carrying blaKPC or blaNDM genes, facilitating rapid dissemination of meropenem resistance among Klebsiella pneumoniae and other Enterobacterales.[123] CRISPR-AMRtracker and Cas12a-based assays enable sensitive detection and monitoring of carbapenemase gene transfer, supporting global efforts to map resistance hotspots and inform targeted interventions.[124][125] Whole-genome sequencing complements these methods, highlighting the role of plasmids in propagating resistance, with projections indicating sustained spread without enhanced surveillance.[126]Pharmacokinetic enhancements, such as oral prodrugs, are in early development to improve meropenem's bioavailability, which is currently limited to intravenous use due to gastric instability. Prodrug strategies involving lipophilic promoieties have demonstrated increased absorption in preclinical evaluations, potentially enabling outpatient treatment of resistant infections by 2030.[127] These modifications aim to inhibit renal tubular secretion and protect against enzymatic degradation, enhancing overall exposure against carbapenem-resistant strains.[128]NIH funding supports antibiotic stewardship models to curb meropenem resistance, with grants like the Large Research Projects for Combating Antibiotic-Resistant Bacteria initiative allocating resources for implementation studies and resistance tracking.[129] These efforts, aligned with the National Action Plan for Combating Antibiotic-Resistant Bacteria (2020-2025), aim to reduce the impact of AMR through optimized prescribing and surveillance.[130]