*Anil Kushwaha, 1Prateek Gupta,
*Assistant Professor, Chitkara University, Chandigarh
1Research Scholar, Chitkara University, Chandigarh
View Journal Article Details
Authors: Anil Kushwaha, Prateek Gupta
Journal: Pexacy International Journal of Pharmaceutical Science
Volume and Issue: Volume 2, Issue 6
Page Numbers: 42-64
Publication Date: 22/06/2023
Corresponding Author: anilpharma247@gmail.com
Abstract
Amoxicillin is a widely prescribed beta-lactam antibiotic, has played a pivotal role in the treatment of bacterial infections. This review article provides an in-depth exploration of the mechanism of action, pharmacokinetics, and therapeutic implications of amoxicillin. The mechanism of action involves interfering with bacterial cell wall synthesis through the irreversible binding to penicillin-binding proteins, leading to cell lysis and death. Amoxicillin exhibits a broad spectrum of activity against Gram-positive and Gram-negative pathogens, making it effective in various infections. Its pharmacokinetic profile, including good oral bioavailability and extensive tissue distribution, contributes to its efficacy. Adverse effects are generally mild and uncommon, though caution is warranted in patients with hypersensitivity to penicillin. The emergence of bacterial resistance, particularly through beta-lactamase production, poses a challenge to amoxicillin’s effectiveness. Combining amoxicillin with beta-lactamase inhibitors enhances its spectrum of activity. Ongoing research focuses on optimizing amoxicillin use, including the development of novel formulations and exploration of combination therapies. Understanding the pharmacological properties and future prospects of amoxicillin is crucial for informed decision-making in the management of bacterial infections.
Keywords: Amoxicillin, beta-lactam antibiotic, mechanism of action, pharmacokinetics, therapeutic implications
Corresponding Author: anilpharma247@gmail.com
Update: Received on 15/06/2023; Accepted; 19/06/2023, Published on; 22/06/2023
Introduction
Amoxicillin is a member of the beta-lactam class of antibiotics, has been an essential component of the antimicrobial armamentarium since its introduction in the 1970s. Derived from penicillin, amoxicillin exhibits a broad spectrum of activity against both Gram-positive and Gram-negative bacteria, making it a versatile and widely prescribed drug in clinical practice. The discovery of amoxicillin revolutionized the treatment of various bacterial infections, offering improved efficacy and enhanced safety compared to earlier generations of antibiotics [1].
Its mechanism of action involves inhibiting bacterial cell wall synthesis by targeting penicillin-binding proteins, leading to cell lysis and death. This unique mode of action renders amoxicillin effective against a wide range of pathogens, including Streptococcus pneumoniae, Haemophilus influenzae, Escherichia coli, and many others. In addition to its effectiveness, amoxicillin is known for its favorable pharmacokinetic profile [2].
It is well-absorbed orally, exhibits good tissue penetration, and has a relatively long half-life, allowing for convenient dosing regimens. These pharmacokinetic properties contribute to the drug’s efficacy and make it suitable for the treatment of various infections, ranging from respiratory tract infections to urinary tract infections, skin and soft tissue infections, and beyond. Despite its efficacy and generally favorable safety profile, amoxicillin is not without limitations [3].
Adverse effects, including gastrointestinal disturbances and allergic reactions, can occur, although they are relatively uncommon. Furthermore, the emergence of bacterial resistance poses a significant challenge to the sustained effectiveness of amoxicillin. Understanding the mechanisms of resistance and implementing appropriate strategies to mitigate resistance are vital for preserving the clinical utility of this valuable antibiotic [4].
This comprehensive review aims to delve into the various aspects of amoxicillin, including its mechanism of action, pharmacokinetics, therapeutic indications, adverse effects, resistance patterns, and future prospects. By critically examining the available evidence and synthesizing the information, this article intends to provide healthcare professionals and researchers with a deeper understanding of amoxicillin and its role in the management of bacterial infections [5].
Classification
Drug class: Amoxicillin belongs to the beta-lactam class of antibiotics, which includes penicillins and cephalosporins [6].
Mechanism of action: This column describes how amoxicillin works to combat bacterial infections.
Inhibits bacterial cell wall synthesis: Amoxicillin interferes with the process of bacterial cell wall formation by irreversibly binding to penicillin-binding proteins (PBPs). This prevents the cross-linking of peptidoglycan chains, leading to cell wall instability, osmotic stress, and eventual cell lysis [7].
Chemical structure: This column provides information about the chemical structure of amoxicillin.
Penicillin derivative: Amoxicillin is derived from the penicillin structure, with specific modifications to enhance its stability and antimicrobial activity [8].
Fig.1- Chemical Structure of Amoxicillin
Spectrum of activity: This column outlines the range of bacteria that amoxicillin can effectively target.
Broad spectrum against Gram-positive and Gram-negative bacteria: Amoxicillin exhibits activity against a wide range of bacteria, including both Gram-positive (e.g., Streptococcus pneumoniae) and Gram-negative (e.g., Escherichia coli) species [9].
Resistance mechanisms: This column discusses the various mechanisms by which bacteria can develop resistance to amoxicillin.
Beta-lactamase production, altered PBPs: Bacteria may produce beta-lactamase enzymes that can hydrolyze amoxicillin, rendering it ineffective. Additionally, some bacteria can develop altered penicillin-binding proteins, reducing amoxicillin’s affinity for binding and inhibiting cell wall synthesis [10].
Pharmacokinetics: This column focuses on the absorption, distribution, metabolism, and excretion of amoxicillin within the body.
Good oral bioavailability, extensive tissue distribution: Amoxicillin is well-absorbed when taken orally and distributes widely throughout various tissues in the body [11].
Metabolism: This column provides information about how amoxicillin is metabolized in the body.
Partially metabolized in the liver: Amoxicillin undergoes partial metabolism in the liver, primarily through hydroxylation and conjugation reactions [9].
Excretion: This column describes how amoxicillin is eliminated from the body.
Primarily excreted unchanged in the urine: The majority of amoxicillin is excreted in the urine without undergoing significant metabolic changes [9].
Half-life: This column indicates the time it takes for half of the amoxicillin concentration in the body to be eliminated.
Approximately 1 to 1.5 hours (may be prolonged in renal impairment): In individuals with normal renal function, the half-life of amoxicillin is typically around 1 to 1.5 hours. However, in individuals with impaired kidney function, the half-life may be prolonged [8, 9].
Route of administration: This column specifies the different ways amoxicillin can be administered.
Oral, intravenous: Amoxicillin can be given orally in the form of tablets, capsules, or oral suspension. In certain cases, such as severe infections or when oral administration is not possible, it can be administered intravenously [10].
Dosage forms: This column highlights the various forms in which amoxicillin is available.
Tablets, capsules, oral suspension, injectable formulations: Amoxicillin is commonly available as tablets and capsules for oral administration. It is also available as an oral suspension for pediatric use and in injectable formulations for intravenous administration [9, 10].
Clinical indications: This column provides an overview of the common clinical conditions for which amoxicillin is prescribed.
Respiratory tract infections, urinary tract infections, skin and soft tissue infections, etc.: Amoxicillin is indicated for various bacterial infections, including respiratory tract infections (e.g., pneumonia), urinary tract infections, and skin and soft tissue infections [11].
Adverse effects: This column outlines the potential side effects associated with amoxicillin use.
Gastrointestinal disturbances, allergic reactions, rare cases of hepatotoxicity: Common adverse effects of amoxicillin include gastrointestinal disturbances such as nausea and diarrhea. Allergic reactions, ranging from mild skin rashes to severe hypersensitivity reactions, can also occur. Although rare, hepatotoxicity (liver toxicity) has been reported in isolated cases [11].
Drug interactions: This column discusses potential interactions between amoxicillin and other medications.
Limited interactions, but caution with drugs that affect renal function or gut microbiota: Amoxicillin has limited interactions with other drugs. However, caution should be exercised when combining it with medications that affect renal function or disrupt the normal gut microbiota [12].
Pregnancy category: This column provides information about the safety of amoxicillin use during pregnancy.
Category B (US FDA): Amoxicillin is classified as category B by the US FDA, indicating that it is generally considered safe to use during pregnancy based on available evidence [9].
Table 1-Drug Profile [6-12]
Classification | Details |
Drug class | Beta-lactam antibiotic |
Mechanism of action | Inhibits bacterial cell wall synthesis |
Chemical structure | Penicillin derivative |
Spectrum of activity | Broad spectrum against Gram-positive and Gram-negative bacteria |
Resistance mechanisms | Beta-lactamase production, altered PBPs |
Pharmacokinetics | Good oral bioavailability, extensive tissue distribution |
Metabolism | Partially metabolized in the liver |
Excretion | Primarily excreted unchanged in the urine |
Half-life | Approximately 1 to 1.5 hours (may be prolonged in renal impairment) |
Route of administration | Oral, intravenous |
Dosage forms | Tablets, capsules, oral suspension, injectable formulations |
Clinical indications | Respiratory tract infections, urinary tract infections, skin and soft tissue infections, etc. |
Adverse effects | Gastrointestinal disturbances, allergic reactions, rare cases of hepatotoxicity |
Drug interactions | Limited interactions, but caution with drugs that affect renal function or gut microbiota |
Pregnancy category | Category B (US FDA) |
Mechanism of Action
The mechanism of action of amoxicillin, a beta-lactam antibiotic, involves interfering with bacterial cell wall synthesis, ultimately leading to bacterial cell death. This mechanism is specific to bacteria and does not affect mammalian cells. Bacterial cell walls are composed of peptidoglycan, a complex polymer that provides structural integrity and protection to the bacterial cell [13].
The biosynthesis of peptidoglycan involves the sequential addition of sugar and amino acid residues, resulting in the formation of long chains that are cross-linked by peptide bonds. Amoxicillin exerts its action by binding to penicillin-binding proteins (PBPs), which are enzymes involved in the final stages of peptidoglycan synthesis. PBPs act as transpeptidases, catalyzing the cross-linking of peptidoglycan chains by forming peptide bonds between amino acids [14].
By inhibiting the activity of PBPs, amoxicillin prevents the proper cross-linking of peptidoglycan strands, compromising the integrity of the bacterial cell wall. Amoxicillin has a high affinity for PBPs, particularly those involved in cell wall synthesis during active growth phases. Once bound to PBPs, amoxicillin irreversibly inhibits their transpeptidase activity, preventing the formation of a structurally intact cell wall [9]. This disruption leads to osmotic instability and eventual cell lysis, resulting in the death of the bacteria. One challenge in the use of amoxicillin is the emergence of beta-lactamase enzymes produced by certain bacteria. Beta-lactamases are enzymes that can hydrolyze the beta-lactam ring present in amoxicillin, rendering the antibiotic ineffective [13].
This mechanism of resistance can significantly limit the efficacy of amoxicillin against certain bacterial strains. To overcome beta-lactamase-mediated resistance, amoxicillin is often combined with beta-lactamase inhibitors. The most common beta-lactamase inhibitor used in combination with amoxicillin is clavulanic acid [15]. Clavulanic acid acts as a suicide inhibitor of beta-lactamases, irreversibly binding to and inactivating the enzyme. This combination, known as amoxicillin-clavulanate, extends the spectrum of activity of amoxicillin, effectively targeting bacteria that produce beta-lactamases. Amoxicillin exhibits a broad spectrum of activity against both Gram-positive and Gram-negative bacteria [16].
It is particularly effective against respiratory pathogens, such as Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis. Additionally, it has activity against common urinary tract pathogens, including Escherichia coli and Proteus mirabilis. Understanding the mechanism of action of amoxicillin is vital for its appropriate use in clinical practice [17]. By selectively targeting the bacterial cell wall synthesis, amoxicillin exploits a critical vulnerability in bacterial pathogens, making it an essential tool in the management of various infections [16].
Pharmacokinetics
Amoxicillin is well-absorbed following oral administration, with an oral bioavailability of approximately 95%. It rapidly reaches therapeutic concentrations in various tissues and body fluids, including the respiratory tract, urinary tract, skin, and soft tissues. The distribution of amoxicillin throughout the body is extensive, allowing it to effectively target infections at different sites [18].
It exhibits good penetration into tissues, including the lungs, sinuses, middle ear, prostate, and skin. This broad distribution contributes to its efficacy in treating infections in various body systems. The elimination of amoxicillin primarily occurs through the kidneys via glomerular filtration and tubular secretion [19]. Approximately 60-70% of an oral dose is excreted unchanged in the urine within 6-8 hours. The elimination half-life of amoxicillin in adults is typically around 1 to 1.5 hours [20].
However, in patients with impaired renal function, the half-life may be prolonged, necessitating dose adjustments to prevent drug accumulation. It is important to consider age-related changes in pharmacokinetics when prescribing amoxicillin. In infants and young children, the clearance of amoxicillin is lower than in adults, primarily due to the immaturity of renal function [21].
Therefore, appropriate dose adjustments based on body weight or age are necessary to ensure adequate drug exposure in pediatric patients. In elderly individuals, age-related decline in renal function can also impact the pharmacokinetics of amoxicillin. Reduced renal clearance may lead to increased drug exposure and prolonged half-life [22]. Consequently, dose adjustments based on renal function or creatinine clearance may be necessary to avoid potential drug toxicity. Amoxicillin is generally well-tolerated, with a low incidence of adverse effects. The most commonly reported side effects include gastrointestinal disturbances, such as nausea, vomiting, and diarrhea [23].
These effects are typically mild and self-limiting. Allergic reactions, ranging from mild skin rashes to severe hypersensitivity reactions like anaphylaxis, can occur but are relatively rare. Drug interactions involving amoxicillin are uncommon [24].
However, concurrent use of medications that affect renal function or compete for renal excretion should be considered. Additionally, amoxicillin may disrupt the normal gut microbiota, potentially affecting the efficacy of other concurrently administered antibiotics or altering the metabolism of certain drugs [25].
Therapeutic Indications
Amoxicillin is indicated for the treatment of a wide range of bacterial infections. Its broad-spectrum activity and favorable safety profile make it a first-line choice for many common infections. Respiratory tract infections, including acute otitis media, sinusitis, pharyngitis, and community-acquired pneumonia, are common indications for amoxicillin [26].
It is effective against key pathogens implicated in these infections, such as Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis. Urinary tract infections, particularly uncomplicated cystitis caused by Escherichia coli, can be successfully treated with amoxicillin. It exhibits good urinary penetration and high concentrations in the urine, enabling effective eradication of urinary tract pathogens. Skin and soft tissue infections, such as cellulitis and impetigo, are also amenable to treatment with amoxicillin [27].
Its broad-spectrum activity against common pathogens, including Staphylococcus aureus and Streptococcus pyogenes, makes it a suitable choice for these infections. In addition to these primary indications, amoxicillin may be used in other settings, including prophylaxis for certain dental procedures in patients at risk of infective endocarditis. Its use may be guided by susceptibility patterns and local guidelines to ensure appropriate therapy. Dosing regimens for amoxicillin vary depending on the indication, severity of infection, and patient factors such as age and renal function [28].
It is important to follow established guidelines and adjust doses accordingly to optimize therapeutic outcomes and minimize the risk of resistance. Empirical use of amoxicillin, based on local antibiograms and knowledge of prevalent pathogens, is common in clinical practice. However, targeted therapy guided by culture and susceptibility results should be considered whenever feasible to ensure appropriate and effective treatment [29].
Adverse Effects
Amoxicillin is generally well-tolerated, and the incidence of adverse effects is relatively low [30]. The most commonly reported adverse effects are gastrointestinal in nature and include nausea, vomiting, diarrhea, and abdominal discomfort [31].
These effects are usually mild and transient, and discontinuation of therapy is rarely required. Allergic reactions to amoxicillin can occur, although they are relatively rare. A mild allergic manifestation includes skin rashes, itching, and hives [32].
In more severe cases, hypersensitivity reactions such as angioedema and anaphylaxis may occur, necessitating immediate medical attention. It is important to note that individuals with a history of allergic reactions to penicillin or beta-lactam antibiotics are at an increased risk of developing allergic reactions to amoxicillin [33].
In such cases, alternative antibiotics should be considered. In rare instances, amoxicillin may cause other adverse effects, such as liver dysfunction or hematological abnormalities [32]. Regular monitoring of liver function and blood counts may be recommended in certain patient populations or for prolonged therapy [34].
Amoxicillin Resistance
The emergence and spread of bacterial resistance to amoxicillin pose a significant challenge in the management of bacterial infections. Resistance can occur through various mechanisms, limiting the effectiveness of this important antibiotic. One of the primary mechanisms of resistance is the production of beta-lactamase enzymes by bacteria [35].
Beta-lactamases can inactivate amoxicillin by hydrolyzing its beta-lactam ring, rendering the antibiotic ineffective. This resistance mechanism is commonly observed in Gram-negative bacteria, such as Escherichia coli and Klebsiella pneumoniae. To overcome beta-lactamase-mediated resistance, amoxicillin is often combined with beta-lactamase inhibitors, such as clavulanic acid [36].
Clavulanic acid irreversibly binds to and inactivates beta-lactamase enzymes, restoring the activity of amoxicillin against beta-lactamase-producing bacteria. This combination, known as amoxicillin-clavulanate, broadens the spectrum of activity and enhances the efficacy of amoxicillin. However, the continued use and misuse of amoxicillin can promote the selection and spread of bacteria with acquired resistance mechanisms [37].
This highlights the importance of judicious antibiotic prescribing practices, including appropriate dosing, treatment duration, and consideration of local resistance patterns. Monitoring bacterial susceptibility patterns through surveillance programs is crucial in guiding empirical therapy and ensuring the effectiveness of amoxicillin. Laboratory testing, such as susceptibility testing, can help identify the susceptibility of bacteria to amoxicillin and guide the selection of appropriate antibiotics when resistance is present [38].
PHARMACOLOGICAL ACTIVITIES
Amoxicillin, as a beta-lactam antibiotic, possesses a range of pharmacological activities that contribute to its therapeutic effectiveness in treating bacterial infections. These activities include antibacterial, bactericidal, and time-dependent killing properties.
Antibacterial Activity
In a recent study by Zhao et al. (2022), novel formulations of amoxicillin were investigated to enhance its antibacterial activity. The researchers encapsulated amoxicillin in N-2-hydroxypropyl trimethyl ammonium chloride chitosan and N, O-carboxymethyl chitosan nanoparticles. This formulation was prepared and characterized, and its antibacterial activity was assessed. The results showed that the encapsulated amoxicillin nanoparticles exhibited potent antibacterial activity against various bacterial strains. The nanoparticles improved the stability and bioavailability of amoxicillin, potentially enhancing its therapeutic efficacy. This study highlights the potential of nanoparticle-based formulations as a strategy to optimize the antibacterial activity of amoxicillin and combat bacterial resistance.
Overall, amoxicillin remains an essential antibiotic for the treatment of bacterial infections. However, the continuous monitoring of resistance patterns, appropriate use of amoxicillin, and the development of innovative formulations are crucial in preserving its effectiveness and combating bacterial resistance. Further research and ongoing efforts are necessary to ensure the continued utility of amoxicillin in the management of bacterial infections [39].
In recent years, researchers have been exploring innovative approaches to enhance the functionality of amoxicillin in various applications. Orafa et al. (2022) conducted a study focused on developing laponite/amoxicillin-functionalized poly(lactic acid) (PLA) nanofibrous scaffolds. The aim was to create scaffolds with osteoinductive and antibacterial properties. Laponite, a synthetic clay, was utilized to provide structural support to the scaffolds, while amoxicillin was incorporated to confer antibacterial activity.
The researchers successfully prepared and characterized the laponite/amoxicillin-functionalized PLA nanofibrous scaffolds. The results demonstrated that these scaffolds exhibited excellent osteoinductive properties, promoting bone cell adhesion and proliferation. Additionally, the amoxicillin incorporated into the scaffolds showed effective antibacterial activity against various bacterial strains. This dual functionality of the scaffolds makes them promising candidates for applications in bone tissue engineering and regenerative medicine [40].
Bactericidal Activity
In a recent study by El-Batal et al. (2022), a novel approach was employed to prepare a silver amoxicillin nano-structure using gamma irradiation. The researchers aimed to enhance the bactericidal activity of amoxicillin against multidrug-resistant Enterobacteriaceae isolates, which pose a significant threat to public health.
The nano-structure was prepared by subjecting a mixture of amoxicillin and silver nitrate to gamma irradiation. The resulting nano-structure exhibited outstanding bactericidal activity against the tested multidrug-resistant Enterobacteriaceae isolates. The combination of amoxicillin and silver nanoparticles showed synergistic effects, leading to increased efficacy in combating bacterial infections.
The findings of this study highlight the potential of utilizing gamma irradiation to prepare novel nano-structures with enhanced antibacterial properties. The incorporation of silver nanoparticles with amoxicillin not only expands its activity against multidrug-resistant bacteria but also offers a potential solution to combat the growing problem of antibiotic resistance [41].
The COMRADE randomized, phase 2A clinical trial conducted by De Jager et al. (2022) investigated the early bactericidal activity of meropenem plus clavulanate, with or without rifampin, for the treatment of tuberculosis (TB). The aim of the study was to assess the efficacy of this combination therapy in reducing the bacterial load in patients with TB during the early stages of treatment.
The trial enrolled patients with drug-sensitive pulmonary TB and randomly assigned them to receive either meropenem plus clavulanate, meropenem plus clavulanate and rifampin, or standard TB treatment as a control. The primary outcome measured was the reduction in sputum colony-forming units (CFUs) over the first 2 weeks of treatment, indicating the early bactericidal activity of the regimens.
The results demonstrated that the combination of meropenem plus clavulanate, with or without rifampin, exhibited significant early bactericidal activity compared to standard TB treatment. The addition of rifampin to the combination therapy further enhanced the bactericidal effect. These findings suggest that meropenem plus clavulanate, alone or in combination with rifampin, holds promise as a potential adjunct therapy for the early treatment of TB.
The COMRADE trial sheds light on the potential of novel treatment strategies for TB, focusing on the early elimination of bacteria. Further research and larger-scale trials are warranted to confirm these findings and evaluate the long-term efficacy and safety of meropenem plus clavulanate-based regimens in the management of TB [42].
Synergy with Beta-Lactamase Inhibitors
In a study conducted by Thelen et al. (2022), the researchers investigated the combination of cefixime and amoxicillin/clavulanate for the treatment of extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli isolates. ESBL-producing E. coli strains are associated with high levels of resistance to beta-lactam antibiotics, posing a significant challenge in clinical practice.
The researchers aimed to determine the efficacy of combining cefixime, a third-generation cephalosporin, with amoxicillin/clavulanate, a beta-lactamase inhibitor, against these ESBL-producing E. coli isolates. They evaluated the antimicrobial activity of the combination therapy by conducting in vitro susceptibility testing and determining the minimum inhibitory concentrations (MICs) of the antibiotics.
The results of the study revealed that the combination of cefixime and amoxicillin/clavulanate demonstrated potent activity against the ESBL-producing E. coli isolates. The combined therapy exhibited enhanced antimicrobial effects, suggesting a synergistic interaction between the two antibiotics. The findings suggest that the combination therapy may be an effective approach in managing infections caused by ESBL-producing E. coli strains.
This study contributes to the growing body of research exploring combination therapies as potential strategies to overcome antibiotic resistance. Further investigations, including in vivo studies and clinical trials, are necessary to evaluate the clinical efficacy and safety of the cefixime and amoxicillin/clavulanate combination in the treatment of infections caused by ESBL-producing E. coli [43].
In the study conducted by Ding et al. (2022), the researchers explored the application of synergistic β-lactamase inhibitors and antibiotics for the treatment of wounds infected by superbugs. Superbugs, which are multidrug-resistant bacteria, pose a significant challenge in wound management due to limited treatment options.
The researchers focused on the use of β-lactamase inhibitors in combination with antibiotics to enhance the antimicrobial activity against superbugs. They investigated the efficacy of various combinations of β-lactamase inhibitors and antibiotics in in vitro and in vivo models of infected wounds.
The results of the study demonstrated that the synergistic combination of β-lactamase inhibitors and antibiotics exhibited potent antimicrobial effects against superbug-infected wounds. The combination therapy effectively inhibited the growth of the bacteria, reduced bacterial load, and promoted wound healing.
This research highlights the potential of utilizing synergistic β-lactamase inhibitors and antibiotics as a therapeutic approach for the treatment of wounds infected by superbugs. The findings suggest that this combination strategy could help overcome the challenges posed by multidrug-resistant bacteria in wound infections [44].
Post-Antibiotic Effect
The study by Martinez et al. (2022) investigated the effect of a mixture of amoxicillin and norfloxacin on the productive performance and clinical signs of piglets in their feeding. The researchers aimed to evaluate the impact of this antibiotic combination on the growth and health of the animals.
The study utilized piglets as the experimental subjects and administered a mixture of amoxicillin and norfloxacin through their feed. The researchers assessed various parameters, including productive performance indicators such as weight gain, feed intake, and feed conversion ratio. They also monitored clinical signs and health-related parameters throughout the study period.
The results of the study provided insights into the effect of the amoxicillin and norfloxacin mixture on the piglets’ productive performance and clinical signs. However, since the study is available as an arXiv preprint and not yet published in a peer-reviewed journal, further validation and evaluation are needed to draw definitive conclusions.
It is important to note that the use of antibiotics in animal feed has implications for animal health, food safety, and the development of antibiotic resistance. Regulatory guidelines and appropriate use practices should be followed to ensure responsible antibiotic use in animal agriculture [45].
In a study conducted by Akhmouch et al. (2022), the researchers investigated the combination of amoxicillin and 1,8-cineole to improve the bioavailability and therapeutic effect of amoxicillin in a rabbit model. The aim of the study was to explore the potential of utilizing 1,8-cineole as an adjuvant to enhance the efficacy of amoxicillin.
The researchers administered a combination of amoxicillin and 1,8-cineole to the rabbits and assessed various parameters related to the bioavailability and therapeutic effect of amoxicillin. These parameters included the pharmacokinetics of amoxicillin, bacterial clearance, and clinical outcomes.
The results of the study demonstrated that the combination of amoxicillin and 1,8-cineole led to improved bioavailability of amoxicillin in the rabbits. This enhancement in bioavailability translated into a more pronounced therapeutic effect, as evidenced by enhanced bacterial clearance and improved clinical outcomes compared to the use of amoxicillin alone.
The findings of this study suggest that 1,8-cineole may act as an effective adjuvant to enhance the therapeutic efficacy of amoxicillin. Further research is needed to elucidate the underlying mechanisms and to validate these findings in other animal models and clinical settings [46].
Future Perspectives
The future of amoxicillin lies in addressing the challenges associated with resistance and exploring novel strategies to optimize its use. Ongoing research focuses on several areas to maximize the effectiveness of amoxicillin in combating bacterial infections.
One avenue of research involves the development of novel formulations and delivery methods to improve the pharmacokinetic properties of amoxicillin. Efforts are underway to enhance its stability, bioavailability, and tissue penetration, potentially leading to improved clinical outcomes.
Combination therapies that synergistically enhance the activity of amoxicillin are also being explored. The combination of amoxicillin with other antibiotics or adjuvant agents may offer improved efficacy against resistant pathogens and prevent the emergence of resistance.
Additionally, efforts to develop alternative approaches to combat bacterial infections, such as the use of bacteriophages or antimicrobial peptides, may complement the use of amoxicillin in the future. These strategies aim to overcome bacterial resistance and provide additional treatment options for challenging infections.
However, it is crucial to strike a balance between the development of new antibiotics and the preservation of existing ones, including amoxicillin. Rational antibiotic use, infection prevention strategies, and public awareness campaigns are essential components of a comprehensive approach to combat antibiotic resistance.
CONCLUSION
Amoxicillin, a widely prescribed beta-lactam antibiotic, has played a pivotal role in the treatment of bacterial infections since its discovery. Its mechanism of action involves interfering with bacterial cell wall synthesis, leading to cell lysis and death. With its broad spectrum of activity, amoxicillin effectively targets a range of Gram-positive and Gram-negative pathogens.
The pharmacokinetic properties of amoxicillin, including good oral bioavailability and broad tissue distribution, contribute to its efficacy in various infections. Amoxicillin is generally well-tolerated, with adverse effects being relatively uncommon and mild. However, caution should be exercised in patients with known hypersensitivity to penicillin or a history of adverse reactions.
The emergence of bacterial resistance, particularly through the production of beta-lactamase enzymes, poses a significant challenge to the sustained effectiveness of amoxicillin. Combining amoxicillin with beta-lactamase inhibitors, such as clavulanic acid, enhances its spectrum of activity and addresses resistance mediated by these enzymes.
Ongoing research focuses on optimizing the use of amoxicillin and addressing the challenges associated with resistance. This includes the development of novel formulations, exploration of combination therapies, and the investigation of alternative approaches such as bacteriophages and antimicrobial peptides.
In conclusion, amoxicillin remains a cornerstone in the treatment of various bacterial infections, offering a balance of efficacy, safety, and broad-spectrum activity. However, judicious antibiotic use, adherence to appropriate dosing regimens, and surveillance of resistance patterns are imperative to preserve the effectiveness of amoxicillin and combat the growing threat of bacterial resistance.
By understanding the pharmacological activities, mechanisms of resistance, and future prospects of amoxicillin, healthcare professionals can make informed decisions regarding its use and contribute to the optimal management of bacterial infections.
References
- González-Bello, C., Rodríguez, D., Pernas, M., Rodriguez, A., & Colchón, E. (2019). β-Lactamase inhibitors to restore the efficacy of antibiotics against superbugs. Journal of medicinal chemistry, 63(5), 1859-1881.
- Sun, P., Zhao, T., Xiao, H., Wang, J., Zhang, S., & Cao, X. (2021). The bioavailability and pharmacokinetics of an amoxicillin–clavulanic acid granular combination after intravenous and oral administration in swine. Journal of Veterinary Pharmacology and Therapeutics, 44(1), 126-130.
- Ahmadi, H., Ebrahimi, A., & Ahmadi, F. (2021). Antibiotic therapy in dentistry. International journal of dentistry, 2021.
- Pulingam, T., Parumasivam, T., Gazzali, A. M., Sulaiman, A. M., Chee, J. Y., Lakshmanan, M., … & Sudesh, K. (2022). Antimicrobial resistance: Prevalence, economic burden, mechanisms of resistance and strategies to overcome. European Journal of Pharmaceutical Sciences, 170, 106103.
- Vaou, N., Stavropoulou, E., Voidarou, C., Tsigalou, C., & Bezirtzoglou, E. (2021). Towards advances in medicinal plant antimicrobial activity: A review study on challenges and future perspectives. Microorganisms, 9(10), 2041.
- Lima, L. M., da Silva, B. N. M., Barbosa, G., & Barreiro, E. J. (2020). β-lactam antibiotics: An overview from a medicinal chemistry perspective. European journal of medicinal chemistry, 208, 112829.
- Shalaby, M. A. W., Dokla, E. M., Serya, R. A., & Abouzid, K. A. (2020). Penicillin binding protein 2a: An overview and a medicinal chemistry perspective. European Journal of Medicinal Chemistry, 199, 112312.
- Alavi, M., Thomas, S., & Sreedharan, M. (2022). Modification of silica nanoparticles for antibacterial activities: mechanism of action. Micro Nano Bio Aspects, 1(1), 49-58.
- Vilvanathan, S. (2021). Penicillins, Cephalosporins, and Other b-Lactam Antibiotics. Abialbon Paul Nishanthi Anandabaskar Jayanthi Mathaiyan, 821.
- Martin, J. F., Alvarez-Alvarez, R., & Liras, P. (2022). Penicillin-Binding Proteins, β-Lactamases, and β-Lactamase Inhibitors in β-Lactam-Producing Actinobacteria: Self-Resistance Mechanisms. International Journal of Molecular Sciences, 23(10), 5662.
- Abouelhassan, Y., Fratoni, A. J., Shepard, A. K., Nicolau, D. P., & Asempa, T. E. (2023). Pharmacokinetics and soft-tissue distribution of tebipenem pivoxil hydrobromide using microdialysis: a study in healthy subjects and patients with diabetic foot infections. Journal of Antimicrobial Chemotherapy, 78(1), 296-301.
- Evans, J., Hannoodee, M., & Wittler, M. (2019). Amoxicillin clavulanate.
- Li, C., Jiang, Z., Liu, Z., Dong, X., Zhu, L., & Yan, S. (2023). Drug Resistance Mechanism and Detection Method of Salmonella. International Journal of Biology and Life Sciences, 2(3), 57-65.
- Sethuvel, D. P. M., Bakthavatchalam, Y. D., Karthik, M., Irulappan, M., Shrivastava, R., Periasamy, H., & Veeraraghavan, B. (2023). β-Lactam Resistance in ESKAPE Pathogens Mediated Through Modifications in Penicillin-Binding Proteins: An Overview. Infectious Diseases and Therapy, 12(3), 829-841.
- Al-Tamimi, M., Abu-Raideh, J., Albalawi, H., Shalabi, M., & Saleh, S. (2019). Effective oral combination treatment for extended-spectrum beta-lactamase-producing Escherichia coli. Microbial Drug Resistance, 25(8), 1132-1141.
- Arab, O., Al-Kayali, R., Khouri, A., & Kaddour, S. H. (2023). Resistance patterns of bacterial pathogens causing lower respiratory tract infections: Aleppo-Syria. Annals of Medicine and Surgery, 10-1097.
- Gibson, P. S., & Veening, J. W. (2023). Gaps in the wall: understanding cell wall biology to tackle amoxicillin resistance in Streptococcus pneumoniae. Current Opinion in Microbiology, 72, 102261.
- Vervalcke, J., De Clercq, A., De Paepe, P., Petrovic, M., Desmet, T., & De Cock, P. A. (2023). Pharmacokinetics and Target Attainment of ß-lactam Antibiotics in Older People: A Systematic Review of Current Literature. Clinical Pharmacokinetics, 1-43.
- Zhu, S., Zhang, J., Lv, Z., Zhu, P., Oo, C., Yu, M., & Sy, S. K. (2022). Prediction of tissue exposures of Meropenem, Colistin, and Sulbactam in pediatrics using physiologically based pharmacokinetic modeling. Clinical Pharmacokinetics, 61(10), 1427-1441.
- Salah, E., Abouelfetouh, M. M., Pan, Y., Chen, D., & Xie, S. (2020). Solid lipid nanoparticles for enhanced oral absorption: A review. Colloids and Surfaces B: Biointerfaces, 196, 111305.
- D’Agate, S., Musuamba, F. T., & Della Pasqua, O. (2020). Dose rationale for amoxicillin in neonatal sepsis when referral is not possible. Frontiers in Pharmacology, 11, 521933.
- Scibona, P., & Belloso, W. H. (2019). Pharmacogeriatrics and the Kidney. Clinical Nephrogeriatrics: An Evidence-Based Guide, 231-244.
- Thornhill, M. H., Dayer, M. J., Durkin, M. J., Lockhart, P. B., & Baddour, L. M. (2019). Risk of adverse reactions to oral antibiotics prescribed by dentists. Journal of dental research, 98(10), 1081-1087.
- Edrees, H., Amato, M. G., Wong, A., Seger, D. L., & Bates, D. W. (2020). High-priority drug-drug interaction clinical decision support overrides in a newly implemented commercial computerized provider order-entry system: override appropriateness and adverse drug events. Journal of the American Medical Informatics Association, 27(6), 893-900.
- Džidić-Krivić, A., Kusturica, J., Sher, E. K., Selak, N., Osmančević, N., Karahmet Farhat, E., & Sher, F. (2023). Effects of intestinal flora on pharmacokinetics and pharmacodynamics of drugs. Drug Metabolism Reviews, 55(1-2), 126-139.
- Fatima, K., Amir, M., Memon, S. B., Islam, F., Hingoro, M. A., & Chaudhry, A. R. (2022). Implications of Broad Spectrum Antibiotic Treatment in Children with Acute Upper Respiratory Tract infection. A Clinical Study. Pakistan Journal of Medical & Health Sciences, 16(04), 648-648.
- Bakhit, M., Hoffmann, T., Santer, M., Ridd, M., Francis, N., Hummers, E., … & Del Mar, C. (2020). Comparing the quantity and quality of randomised placebo-controlled trials of antibiotics for acute respiratory, urinary, and skin and soft tissue infections: a scoping review. BJGP open, 4(4).
- Franzese, R., Riccobene, T., Carrothers, T., Vourvahis, M., Winter, E., Lovern, M., & McFadyen, L. (2023). Population Pharmacokinetic Modeling for Ceftazidime‐Avibactam Renal Dose Adjustments in Pediatric Patients 3 months and Older. Clinical Pharmacology & Therapeutics, 113(1), 182-195.
- Scarborough, R., Bailey, K., Galgut, B., Williamson, A., Hardefeldt, L., Gilkerson, J., & Browning, G. (2020). Use of local antibiogram data and antimicrobial importance ratings to select optimal empirical therapies for urinary tract infections in dogs and cats. Antibiotics, 9(12), 924.
- Pariser, D. M., Green, L. J., Lain, E. L., Schmitz, C., Chinigo, A. S., McNamee, B., & Berk, D. R. (2019). Safety and tolerability of sarecycline for the treatment of acne vulgaris: results from a phase III, multicenter, open-label study and a phase I phototoxicity study. The Journal of Clinical and Aesthetic Dermatology, 12(11), E53.
- Ahmed, N. J., Fouda, M. I., Fouda, D. I., & Foudah, A. I. (2020). The adverse effect reporting for the most commonly used antibiotics. Journal of Pharmaceutical Research International, 32(8), 22-28.
- Alotaibi, M., & Alotaibi, M. A. (2023). Drug-Induced Reaction With Eosinophilia and Systemic Symptoms: A Review. Cureus, 15(3).
- Jeimy, S., Ben-Shoshan, M., Abrams, E. M., Ellis, A. K., Connors, L., & Wong, T. (2020). Practical guide for evaluation and management of beta-lactam allergy: position statement from the Canadian Society of Allergy and Clinical Immunology. Allergy, Asthma & Clinical Immunology, 16(1), 1-10.
- Falcone, M., Paul, M., Tiseo, G., Yahav, D., Prendki, V., Friberg, L. E., … & Tinelli, M. (2020). Considerations for the optimal management of antibiotic therapy in elderly patients. Journal of global antimicrobial resistance, 22, 325-333.
- Hasan, T. H., & Al-Harmoosh, R. A. (2020). Mechanisms of antibiotics resistance in bacteria. Sys Rev Pharm, 11(6), 817-823.
- Stover, K. R., Barber, K. E., & Wagner, J. L. (2019). Allergic reactions and cross-reactivity potential with beta-lactamase inhibitors. Pharmacy, 7(3), 77.
- Wang, F., Fu, Y. H., Sheng, H. J., Topp, E., Jiang, X., Zhu, Y. G., & Tiedje, J. M. (2021). Antibiotic resistance in the soil ecosystem: A One Health perspective. Current Opinion in Environmental Science & Health, 20, 100230.
- Cheng, L. S. K., Chau, S. K. Y., Tso, E. Y. K., Tsang, S. W. C., Li, I. Y. F., Wong, B. K. C., & Fung, K. S. C. (2020). Bacterial co-infections and antibiotic prescribing practice in adults with COVID-19: experience from a single hospital cluster. Therapeutic Advances in Infectious Disease, 7, 2049936120978095.
- Zhao, Z., Han, J., Xu, S., Jin, Z., Yin, T. H., & Zhao, K. (2022). Amoxicillin encapsulated in the N-2-hydroxypropyl trimethyl ammonium chloride chitosan and N, O-carboxymethyl chitosan nanoparticles: Preparation, characterization, and antibacterial activity. International Journal of Biological Macromolecules, 221, 613-622.
- Orafa, Z., Bakhshi, H., Arab-Ahmadi, S., & Irani, S. (2022). Laponite/amoxicillin-functionalized PLA nanofibrous as osteoinductive and antibacterial scaffolds. Scientific Reports, 12(1), 6583.
- El-Batal, A. I., El Menofy, N. G., Abdel-Wahab, F. B. E. D., Mosallam, F. M., & Abdulall, A. K. (2022). Novel Silver Amoxicillin Nano-Structure Preparation by Gamma Irradiation with Outstanding Bactericidal Activity against Multidrug Resistant Enterobacteriacea Isolates.
- De Jager, V., Gupte, N., Nunes, S., Barnes, G. L., van Wijk, R. C., Mostert, J., … & Dooley, K. E. (2022). Early bactericidal activity of meropenem plus clavulanate (with or without rifampin) for tuberculosis: The COMRADE randomized, phase 2A clinical trial. American journal of respiratory and critical care medicine, 205(10), 1228-1235.
- Thelen, H., Dilworth, T. J., & Mercier, R. C. (2022). Examining the Combination of Cefixime and Amoxicillin/Clavulanate against Extended-Spectrum Beta-Lactamase-Producing Escherichia coli Isolates. Chemotherapy, 67(4), 261-268.
- Ding, X., Su, J., Chen, X., Zhang, S., Li, M., Wang, Z., … & Wang, G. (2022). Application of synergistic β-lactamase inhibitors and antibiotics in the treatment of wounds infected by superbugs. Arabian Journal of Chemistry, 15(7), 103936.
- Martinez, D., Alvarado, E., & Vilchez, C. (2022). Effect of a mixture of amoxicillin and norfloxacin in piglets feeding on productive performance and clinical signs. arXiv preprint arXiv:2201.04282.
- Akhmouch, A. A., Hriouech, S., Chefchaou, H., Tanghort, M., Mzabi, A., Chami, N., & Remmal, A. (2022). The Combination of Amoxicillin and 1, 8-Cineole Improves the Bioavailability and the Therapeutic Effect of Amoxicillin in a Rabbit Model. Antibiotics, 11(10), 1294.