Next Article in Journal
Effectiveness and Safety of GLP-1 Receptor Agonists in Patients with Type 1 Diabetes
Previous Article in Journal
Transcatheter Repair of Tricuspid Valve Regurgitation: A Systematic Review
Previous Article in Special Issue
Group A Streptococcus Infection in Neonatal Population: A Systematic Review of The Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials

Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
J. Clin. Med. 2024, 13(21), 6530; https://doi.org/10.3390/jcm13216530
Submission received: 11 October 2024 / Revised: 28 October 2024 / Accepted: 29 October 2024 / Published: 30 October 2024

Abstract

:
Pseudomonas aeruginosa (Pa) poses a significant threat to individuals with cystic fibrosis (CF), as this bacterium is highly adaptable and resistant to antibiotics. While early-stage Pa infections can often be eradicated with aggressive antibiotic therapy, chronic infections are nearly impossible to eliminate and require treatments that focus on long-term bacterial suppression. Without such suppression, these persistent infections can severely damage the lungs, leading to serious complications and a reduced life expectancy for CF patients. Evidence for a specific treatment regimen for managing Pa infections in CF patients remains limited. This narrative review provides a detailed analysis of antimicrobial therapies assessed in completed phase IV trials, focusing on their safety and efficacy, especially with prolonged use. Key antibiotics, including tobramycin, colistin, meropenem, aztreonam, ceftolozane/tazobactam, ciprofloxacin, and azithromycin, are discussed, emphasizing their use, side effects, and delivery methods. Inhaled antibiotics are preferred for their targeted action and minimal side effects, while systemic antibiotics offer potency but carry risks like nephrotoxicity. The review also explores emerging treatments, such as phage therapy and antibiofilm agents, which show promise in managing chronic infections. Nonetheless, further research is necessary to enhance the safety and effectiveness of existing therapies while investigating new approaches for better long-term outcomes.

1. Introduction

Cystic fibrosis (CF) is an incurable disease that affects the production of healthy mucus. This leads to a buildup of abnormal mucus that damages the lungs, digestive system, and other vital organs [1,2]. The global prevalence of diagnosed CF cases remains challenging to determine due to underdiagnosis limitations. However, the estimates suggest CF cases range from approximately 70,000 to over 160,000 [3,4]. The variations in CF prevalence across studies can be attributed to the heterogeneous approaches implemented by individual countries in their CF-screening programs [5]. This underscores the need for standardized methodologies to investigate CF prevalence on a global scale.
CF is caused by defects in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, following an autosomal recessive pattern of inheritance. An individual inherits two copies of the CFTR gene, one from each parent; however, only those who receive two mutated copies will develop the disease. [6]. Despite the existence of over 2000 different genetic variations in the CFTR gene, around 700 are identified as disease-causing variants [7,8,9]. Moreover, data suggest that the prevalence of CFTR mutation carriers ranges from 4% to 5% in the general population [10,11,12,13].
In a healthy state, the CFTR protein resides in the apical membrane of epithelial cells in organs like the lungs, liver, pancreas, intestines, and sweat glands. It functions as a chloride channel, regulating the movement of ions and water across these cells, maintaining a thin low-viscosity layer of mucus [14,15]. However, in patients with CF (pwCF), the absence or malfunction of the CFTR protein leads to the production of thick, sticky mucus that can damage organs over time, leading to serious health complications [15].
Lungs are the organs most severely affected in pwCF due to impaired mucociliary clearance caused by thickened mucus secretions. This dysfunction traps inhaled debris and pathogens, creating a breeding ground for chronic infections that are often accompanied by a chronic neutrophilic inflammatory response [16,17]. While the inflammatory response is crucial for fighting infection, prolonged neutrophil recruitment and inflammation can contribute to collateral damage in the lung tissues [18]. Neutrophils exacerbate lung damage through destructive degranulation and the release of neutrophil extracellular traps (NETs), which consist of DNA and antimicrobial proteins. Although NETs are intended to trap and eliminate infectious agents, their dysregulation contributes to excessive inflammation and tissue destruction in CF patients. The prolonged presence of neutrophils and their harmful by-products further damages lung tissues, leading to a progressive decline in lung function and worsening disease severity [19].
Pseudomonas aeruginosa (Pa) poses one of the greatest concerns for lung infections in pwCF. While other bacterial threats like Staphylococcus aureus and Haemophilus influenza can also establish infections, particularly in the early stages of life, Pa is particularly dangerous due to its ability to thrive in damaged airways, contribute to progressive lung disease, and resist antibiotic treatment [20,21]. By adulthood, it is estimated that Pa colonizes the airways of up to 80% of CF individuals, significantly impacting their health [22,23,24]. It has been shown that in the chronic state, Pa isolates are predominantly mucoid, often overexpressing alginate, which contributes to the formation of highly structured biofilms [25,26]. Furthermore, within biofilms, a subpopulation of bacterial cells (approximately 1%) enters a dormant state known as persistence, which exhibit neither active growth nor immediate death, even when exposed to high concentrations of antibiotics [27].
The early detection of Pseudomonas infection is crucial for improving the life expectancy of pwCF, as eradication therapy with intensive antibiotics is most successful in newly diagnosed cases [28,29,30]. However, once chronic infection is established, Pa becomes resistant to eradication, leading to more severe complications. Chronic Pa infections significantly accelerate the decline in lung function in pwCF, with estimates suggesting a 5–10% decrease in lung capacity per year. This decline can lead to serious complications, including early death [31,32,33]. To address this, a multi-pronged strategy is typically employed to manage the infection burden and increase life expectancy. This involves antimicrobial treatment to suppress bacterial growth, anti-inflammatory medications to manage chronic lung inflammation, and airway clearance therapies, including CFTR modulators, which enhance chloride transport, improve mucus viscosity, and reduce the frequency and severity of lung infections by preventing bacterial build-up [34,35,36]. A recent study in the US reported a significant reduction in mortality rates, with the median age of death increasing from 24 years in 1999 to 37 years in 2020, a change attributed to the implementation of aggressive multi-approach treatments in pwCF [37].

2. Overview of Phase IV Clinical Trials

Phase IV clinical trials are conducted after a drug or therapy has received regulatory approval and becomes available to the public. These studies, which include post-marketing surveillance (PMS), are crucial for evaluating the long-term safety, effectiveness, and potential adverse effects of treatments in a broader and more diverse patient population than those involved in earlier trial phases [38,39]. Unlike phases I–III, which are performed under controlled conditions with carefully selected participants, phase IV trials examine how therapies perform in real-world settings, offering a deeper insight into their practical applications, potential risks, and broader efficacy (Table 1) [40].
Modifying an approved treatment plan to suit a specific population, such as managing infections in pwCF, qualifies as a phase IV trial [38,39]. For CF patients, who often require prolonged therapy, PMS is particularly important for monitoring the long-term safety and efficacy of treatments [41]. The real-world data collected through phase IV trials enable clinicians to determine whether these therapies maintain their effectiveness over time, reduce the frequency of exacerbations, and ensure an acceptable safety profile for prolonged use. Additionally, these trials can identify emerging issues such as drug resistance and individual variations in response, and allow for the refinement of treatment regimens to improve the long-term outcomes in CF care [42].

3. Therapeutic Candidates at Phase IV for Management of Pa Infection in pwCF

Despite the significant advancements in the management of infections in pwCF, ongoing research remains critical to further enhance both patient longevity and quality of life. The development of safer and more efficacious therapeutic strategies to address pseudomonal infections is paramount, given their persistent role in disease progression and associated complications [43]. This review analyzes antipseudomonal therapies assessed in completed Phase IV trials for the treatment of Pa infections in pwCF, providing valuable insights into the effectiveness and safety of current treatment modalities. Additionally, it aims to identify gaps in existing therapeutic approaches, offering a foundation for optimizing future strategies to better manage Pa infections in the CF population. Data for these clinical trials were sourced from the ClinicalTrials.gov database, which offers detailed information on ongoing and completed studies. A more in-depth discussion of these trials is provided in the sections below (Table 2).

3.1. Tobramycin

Tobramycin is a potent antibiotic that belongs to the aminoglycoside class. It demonstrates bactericidal activity against a broad spectrum of Gram-positive and Gram-negative bacteria including Pa, Escherichia coli, Serratia, Enterobacter, and Klebsiella spp. as well as both penicillinase-producing and non-producing strains of S. aureus [44,45]. Tobramycin works by interfering with protein synthesis machinery in bacteria. It binds to the 16S ribosomal subunit, an essential component of the bacterial ribosome responsible for translating mRNA into proteins. This binding disrupts the ability of bacteria to assemble functional proteins, ultimately resulting in cell death [46].
Following the U.S. Food and Drug Administration (FDA) approval in 1975, tobramycin has been established as a versatile therapeutic agent for managing and treating a diverse range of systemic and ocular infections [47,48,49]. It can be administered through various routes depending on the type of infection being treated, with available formulations for inhalation, injection, and topical ophthalmic application. However, the oral administration of tobramycin is limited due to poor bioavailability caused by efflux via P-glycoprotein pumps in the intestinal brush border [50,51,52].
Inhaled tobramycin has emerged as a key therapeutic tool to combat lung infections in pwCF as it is delivered directly to the airways via nebulization or dry powder inhalers. This localized approach minimizes systemic exposure and associated adverse effects (AEs) compared with intravenous antibiotics [50,53,54,55]. The AEs with long-term intake include nausea, vomiting, diarrhea, balance, and hearing problems and, most importantly, kidney damage with prolonged and/or higher doses [45,56,57].

3.2. Colistin

Colistin is a polycationic antibiotic belonging to a class of antibiotics called polymyxins. Colistin, also known as polymyxin E, was first isolated from the bacterium Bacillus colistinus in Japan during 1947. Notably, it exhibits broad-spectrum bactericidal activity against a diverse range of Gram-negative bacteria, especially those characterized by multidrug resistance (MDR) including Enterobacteriaceae, Pa, and Acinetobacter baumannii [58,59].
Colistin targets the lipopolysaccharides (LPS) structure in the outer membrane of Gram-negative bacteria for their antibacterial effect [58]. Bacterial LPS is made up of three main components: (i) a core oligosaccharide linked to the inner membrane; (ii) an outer polysaccharide chain (O-antigen); and (iii) a lipid A portion embedded in the outer membrane, maintaining its integrity. Hence, the electrostatic exchange between the positively charged Dab (diaminobutyric acid) residue on colistin and the phosphates of lipid A disrupts the bacterial membrane, which leads to cell death due to the escape of essential electrolytes and cellular components [58,60]. Additionally, colistin can also bind to the free molecules of LPS after bacterial lysis and neutralizing their toxic effects [61].
Although colistin received FDA approval in 1959, safety concerns about neurotoxicity and nephrotoxicity led to restrictions on its use in the early 1970s. However, more recently, it has re-emerged as a last-line therapeutic option for treating infections unresponsive to conventional antibiotics. This was driven by its superior effectiveness against MDR pathogens and the rarity of colistin resistance [62,63,64].
Colistin exists in two clinically relevant forms with distinct uses. Colistin sulfate (CS) is the active form and is suitable for oral, inhalation, or topical application. In contrast, colistin methanesulfonate sodium (CMS) is a prodrug that needs conversion in the body to become the active form of colistin. This form is intended for parenteral administration (injection or infusion) and may be given by inhalation. For long-term treatment plans, CMS is preferred because it is less toxic and has fewer unwanted effects than the active form of colistin [65,66,67]. The primary concern of colistin lies in its nephrotoxicity, which can lead to kidney damage. This risk increases with higher doses, longer treatment durations, and pre-existing kidney problems. Other reported side effects include neurotoxicity and gastrointestinal issues like nausea, vomiting, and diarrhea [62,68].

3.3. Meropenem

Meropenem is a carbapenem-class, broad-spectrum beta-lactam antibiotic that is highly effective against a wide variety of Gram-positive and Gram-negative bacterial infections [69]. It exerts a bactericidal effect by hindering the construction of the bacterial cell wall. This occurs when the meropenem binds to penicillin-binding proteins (PBPs), enzymes essential for cross-linking peptidoglycan strands in the cell wall. This binding disrupts the formation of a strong and stable cell wall, ultimately leading to the weakening and death of the bacteria [70,71]. Importantly, unlike its predecessor penicillin, meropenem is resistant to degradation by the chromosomal and extended-spectrum beta-lactamase enzymes produced by bacteria to deactivate beta-lactam antibiotics [72].
In response to growing concerns about antibiotic resistance in the 1990s, meropenem entered clinical use and quickly became a cornerstone in treating serious infections, including pulmonary infections in CF patients [73]. Meropenem comes in one form as a sterile powder for injection and it is typically administered intravenously (IV) by infusion method. This is because the orally administered formulations suffer from instability within the gut, poor permeability across the intestinal wall, and consequently, low bioavailability [74,75,76].
Meropenem is generally well-tolerated but can cause side effects. Common ones include injection site reactions, nausea, vomiting, and diarrhea [73]. Meropenem can also disrupt the gut flora, which can lead to metabolic shifts, and increases gut susceptibility to colonization by unwanted pathogens such as Clostridioides difficile [77,78].

3.4. Ceftolozane/Tazobactam

Ceftolozane/tazobactam (CZ/TAZ) is a synergistic antibiotic combination comprising a fifth-generation cephalosporin (ceftolozane) and a beta-lactamase inhibitor (tazobactam). This combination exerts its bactericidal effect against a wide range of organisms including Pa by inhibiting bacterial cell wall production while simultaneously preventing the beta-lactamase-mediated inactivation of ceftolozane [79,80].
CZ/TAZ has received recent approval by both the FDA and the European Medicines Agency (EMA) under the brand name Zerbaxa for treating certain complicated infections. The targeted infections include hospital-acquired pneumonia, complicated urinary tract infections, and intra-abdominal infections. CZ/TAZ is available in only one form as a sterile powder for injection which demonstrates versatility in treating complex infections. However, CZ/TAZ can be combined with metronidazole when anaerobic bacterial involvement is suspected [81,82,83].
Although CZ/TAZ is generally well-tolerated, it comes with certain safety considerations and potential interactions. Common side effects include injection site reactions, such as pain or irritation, as well as gastrointestinal symptoms like nausea, diarrhea, and headaches. While these are typically mild, more serious adverse effects, including muscle cramps, irregular heart rhythms, or kidney dysfunction, can occur in rare instances. Severe allergic reactions, such as anaphylaxis, are possible, particularly in patients with a history of allergies to cephalosporins, penicillin, or other beta-lactam antibiotics, making it crucial to evaluate any hypersensitivity before beginning treatment. Additionally, the prolonged use of CZ/TAZ may predispose patients to secondary infections, including oral thrush or yeast infections, and it carries a risk of developing a serious C. difficile-associated intestinal condition, characterized by persistent diarrhea and abdominal pain, which may arise during or after treatment [81,82].

3.5. Azithromycin

Azithromycin, a broad-spectrum macrolide antibiotic, is extensively used for treating respiratory infections and specific sexually transmitted infections (STIs). It is a derivative of erythromycin, with enhanced potency against Gram-negative bacteria, particularly Enterobacteriaceae and many Gram-positive organisms [84,85]. Azithromycin exerts its antibacterial effect by selectively binding to the 23S rRNA of the 50S ribosomal subunit in susceptible bacteria. This binding disrupts protein translation, thereby inhibiting bacterial growth and replication [86]. While primarily acting as a bacteriostatic agent, azithromycin can exhibit bactericidal effects at higher doses against certain bacteria like H. influenzae and a number of streptococci members [87,88].
Beyond its antibacterial role, azithromycin is recognized for its significant immunomodulatory effects, which are particularly beneficial in managing chronic respiratory diseases. These effects are mediated through multiple mechanisms, including the regulation of cellular signaling pathways and the suppression of pro-inflammatory cells and mediators [89,90]. Specifically, azithromycin suppresses the activation of nuclear factor kappa B (NF-κB), a key transcription factor involved in the production of inflammatory cytokines like IL-6, IL-8, and TNF-α [91,92]. In addition, it reduces the recruitment and activation of neutrophils and macrophages, thereby dampening excessive inflammatory responses [93,94]. Another key mechanism is its ability to inhibit the production of reactive oxygen species (ROS), which helps mitigate oxidative stress in the airways. Through these pathways, azithromycin not only helps control acute inflammation but also prevents the progression of chronic inflammation and subsequent tissue damage, ultimately improving lung function and reducing the frequency of exacerbations in conditions like cystic fibrosis and bronchiectasis [95,96].
Azithromycin can be administered by both the oral and parenteral routes. It has extended-release formulation coupled with comprehensive tissue and intracellular distribution, allowing once-daily dosing and a more concise treatment regimen compared with other antimicrobial agents [97,98]. Moreover, azithromycin is generally recognized as a well-tolerated antimicrobial agent, with a low incidence of discontinuation due to AEs. It also has a superior safety profile compared with other macrolides such as clarithromycin and erythromycin. Hence, it can be employed for long-term prophylaxis with reported potential for reducing exacerbation frequency, sputum volume, and improving lung function in non-cystic fibrosis bronchiectasis patients [99,100,101].

3.6. Ciprofloxacin

Ciprofloxacin belongs to the second-generation class of fluoroquinolone antibiotics. While it has broad-spectrum activity, it is particularly potent against Gram-negative organisms, including Pa, E. coli, and Klebsiella pneumoniae [102,103]. As a fluoroquinolone, ciprofloxacin functions as a bactericidal agent by inhibiting bacterial DNA gyrase and topoisomerase IV, which are crucial enzymes for bacterial DNA replication and repair. By disrupting these processes, it prevents the unwinding and re-ligation of bacterial DNA, leading to DNA fragmentation and, ultimately, bacterial cell death [104,105].
Ciprofloxacin can be administered orally or intravenously, making it a versatile agent for various clinical conditions. It is indicated for the treatment of multiple infections, including respiratory tract infections such as chronic bronchitis and bacterial pneumonia, as well as urinary tract infections (UTIs), including complicated cases like pyelonephritis, and gastrointestinal infections [106,107,108,109]. Oral formulations are available as oral suspensions and immediate- and extended-release tablets, while IV treatment is used for more severe or complicated infections where higher systemic concentrations are required or when oral administration is not feasible [110].
Ciprofloxacin is generally well-tolerated, but like all fluoroquinolones, it carries specific safety concerns. Common side effects include gastrointestinal disturbances (nausea, diarrhea), headaches, and dizziness. More serious, though less frequent, adverse effects include tendonitis, QT prolongation, and central nervous system (CNS) effects such as seizures, particularly in elderly patients or those with underlying conditions [111,112,113]. However, ciprofloxacin is contraindicated in pediatric patients and pregnant women due to concerns over arthropathy [114,115].

3.7. Aztreonam

Aztreonam is a monobactam antibiotic, part of the beta-lactam family, which is distinguished by its unique single-ring structure. Unlike other beta-lactams, such as penicillin and cephalosporins, its structure allows it to target a narrow range of bacteria. Aztreonam is highly effective against Gram-negative bacteria, including Pa, E. coli, K. pneumoniae, and Enterobacter species. However, it has little to no activity against Gram-positive bacteria or anaerobes, making it a specialized treatment option for Gram-negative infections [116,117].
Aztreonam is indicated for the treatment of a range of infections, including respiratory infections such as pneumonia and bronchiectasis, UTIs, septicemia, skin and soft tissue infections, as well as intra-abdominal and gynecological infections caused by Gram-negative bacteria [118,119,120]. It is typically administered either IV or intramuscularly (IM) for systemic infections, as it is poorly absorbed when taken orally [116]. However, for chronic lung infections, aztreonam is also available in an inhaled form, known as aztreonam lysine (AZLI) [119].
Known for its generally favorable safety profile, aztreonam is well-tolerated by most patients. Common side effects include mild reactions at the injection site, gastrointestinal issues like nausea, vomiting, and diarrhea, and occasional rashes. More serious side effects, such as allergic reactions and elevated liver enzymes, are rare [121,122]. A major advantage of aztreonam is its safe use in individuals with allergies to other beta-lactam antibiotics, such as penicillin and cephalosporins, as cross-reactivity is exceptionally uncommon [116].

4. Treatment Approaches for Management of Pa Infections in pwCF

Antibiotics are a key component of a multifaceted approach to manage infections in pwCF which includes anti-inflammatory therapies, mucolytics, bronchodilators, and CFTR modulators to improve lung function and reduce mucus build-up [36,123]. Around 80% of newly acquired Pseudomonas infections can be eradicated using a combinatorial therapeutic approach involving oral, inhaled, and intravenous antibiotics [124]. However, once the infection transitions to a chronic state, eradication becomes nearly impossible, and treatment focuses primarily on suppressing bacterial load and managing symptoms. The optimal antibiotic regimens, including the best combination of drugs, appropriate dosages, and treatment durations, are still areas of active investigation [125,126,127,128]. To date, no single antibiotic or established combination has been proven to achieve the definitive eradication of Pa in pwCF [129]. The diversity of these approaches reflects the ongoing search for the most effective treatment strategies for this complex and long-lasting condition.

4.1. Systemic Drugs

Systemic antibiotics are often chosen for their ability to achieve high systemic concentrations, making them effective in treating severe or widespread infections [130]. However, while systemic antibiotics can be highly effective in the short term, their long-term use raises concerns, including their potential toxicity and alterations in the pathogenicity of gut microbiota [131,132]. Limited data exist regarding the safety profiles of intravenous (IV) tobramycin and colistin for treating acute pulmonary exacerbations (PEx) in CF patients. Concerningly, IV tobramycin has demonstrated a significantly higher frequency of AEs compared with the two IV-dosing regimens of colistin (NCT02918409). Nephrotoxicity emerged as the most significant risk associated with IV tobramycin treatment, potentially leading to treatment failure and a worsening of the patient’s condition. This was evidenced by a higher incidence in patients receiving IV tobramycin (25%) compared with only 4% in those treated with both colistin regimens combined in this two-week duration study. In fact, nephrotoxicity is a well-recognized AE associated with IV administration of anti-Pseudomonas medications like tobramycin and colistin, as observed in several previous studies [133,134,135,136]. Notably, it has been demonstrated that renal function can recover to pre-treatment levels within 4 weeks after discontinuing tobramycin [137]. In contrast, colistin-induced nephrotoxicity exhibits a dose- and time-dependent relationship, with reported incidence varying between 10% and 70% across multiple studies [138,139,140,141,142]. This variation can also be attributed to the different diagnostic criteria and targeted groups of patients employed by studies [143]. Thus, the systemic administration of antibiotics presents a double-edged sword, necessitating judicious use, especially for chronic conditions.

4.2. Inhaled Drugs

Studies have shown that inhaled antibiotics are successful in reducing Pa density in the airways, improving lung function, and decreasing the frequency of PEx in pwCF with chronic infections [55,144]. Inhaled antibiotics offer a distinct advantage over systemic treatment because of their reduced systemic toxicity and the ability to achieve higher concentrations within the respiratory tract. Despite that, mild to serious AEs can arise in most patients with prolonged use [45,49,145]. In an extended phase IV study focused on the long-term safety of tobramycin inhalation powder (TIP) (NCT01519661 and NCT01775137), the drug demonstrated good tolerability with no unpredictable safety signals identified, and its efficacy was maintained throughout the observation period [146]. The reported AEs were comparable to those observed in other long-term studies from phase III [146,147,148]. Infective PEx was the most common adverse event and the primary reason for study discontinuation with a comparable severity to those in relevant studies [146,147,148]. Cough, the second most common adverse event, was found to decrease in frequency over time. This differentiates this phase IV investigation from previous trials that reported consistently higher rates of cough [146,147,148]. This decrease in cough frequency may be attributed to the improved inhalation techniques by patients, and the effectiveness of TIP in rapidly reducing bacterial load over time. This reduction in bacteria likely leads to a subsequent decrease in neutrophilic airway inflammation, which in turn, reduces sputum production and purulence [149,150,151]. While hemoptysis was also common, most cases were mild and likely linked to the underlying disease, as explained earlier by Thompson et al. (2015) [146,152]. Overall, TIP can be considered a safe and tolerable long-term treatment option for CF patients with established lung disease, although some AEs warrant monitoring. However, limitations inherent to the phase IV open-label, single-arm design necessitate consideration [146]. This is because the design is susceptible to reporting bias, especially for subjective AEs. Furthermore, the lack of a comparator group restricts the ability to definitively assess the treatment’s relative efficacy and safety profile when compared with alternative therapies or placebo.
Following the 1980s’ success of localized delivery of tobramycin, more effective treatments for chronic Pa infections in CF have been introduced via inhalation including colistin, aztreonam, and fluoroquinolones [153]. Nebulizers and dry powder inhalers (DPIs) represent two primary modes of direct drug delivery to the respiratory tract. In a comparative study between TIP and nebulized solutions of tobramycin (TIS) and colistimethate (NCT01844778), TIP administration via the T-326 inhaler demonstrated improved usability, reduced total delivery time, and significantly lower contamination rates compared with nebulizers [154]. Indeed, several studies have demonstrated that the reduced administration time and user-friendliness of inhaler devices positively impacts patient adherence to treatment regimens. This ease of use likely translates to minimized medication-handling errors and improved patient satisfaction with the therapy [155,156,157,158,159]. Moreover, DPIs have minimal risk of microbial contamination as frequent cleaning between uses is unnecessary [160]. In spite of the advantages, DPIs have a number of limitations compared with nebulizers. Certain antibiotics are currently unavailable in DPI formulations, restricting their use in this delivery method. Furthermore, the effective utilization of DPIs often requires a proper inhalation technique, potentially limiting their suitability for young children, older adults, or patients with compromised conditions [161].

4.3. Combination Therapies

While combination therapies offer the potential advantage of broader coverage against resistant bacteria, a careful evaluation of potential side effects and drug interactions is needed [162]. A common regimen involves TIP with oral azithromycin, a treatment prescribed to approximately 75% of patients with chronic lung infections [163]. However, the efficacy of this combination remains unclear. In a phase IV trial intended to investigate the impact of adding azithromycin to a standard TIP regimen (NCT02677701), the co-administration of azithromycin with TIP did not produce a synergistic effect in terms of improving lung function, nor did it result in significant changes in AEs or bacterial load when compared with a placebo [164]. Similarly, Mayer-Hamblett et al. (2018) reported no significant difference in the eradication rate of early Pa infection or clinical outcomes between CF patients receiving tobramycin solution for inhalation (TIS) with azithromycin and those receiving TIS alone [165]. On the other hand, several randomized controlled trials involving azithromycin have demonstrated improvements in lung function, a decrease in PEx and hospitalizations, and a reduced need for intravenous antibiotics in pwCF [166,167,168]. These benefits are likely attributable to the anti-inflammatory properties of azithromycin when used long-term. This can be explained by the shorter duration of a phase IV study (NCT02677701) compared with the other controlled trials and also that the Mayer-Hamblett study (2018) targeted early-stage infections before the establishment of chronic Pa colonization [165]. Therefore, the long-term use of azithromycin could be beneficial in improving lung function as a prophylactic measure, not through direct antimicrobial activity, but by mitigating inflammatory consequences and disease progression. However, more optimized studies are required to confirm these benefits in CF patients.

4.4. Optimal Dosing

Emerging evidence suggests that optimal dosing regimens may differ in CF population compared with non-CF patients [169]. Medication misuse, including overuse, can lead to undesirable side effects, compromising the intended therapeutic benefit and potentially culminating in treatment failure. To address this, two studies (NCT01429259 and NCT02421120) investigated the pharmacokinetics and tolerability of meropenem and ceftolozane/tazobactam (CZ/TAZ) in children and adult CF patients experiencing acute PEx, respectively. Interestingly, meropenem exhibited a higher clearance rate compared with previously reported values in healthy children [170,171,172,173]. In contrast, the findings of CZ/TAZ clearance revealed no significant difference between adult with and without CF condition [174,175,176]. However, the conclusive determination of optimal dosing regimens remains uncertain due to limitations in both phase IV studies. The relatively small sample sizes and the comparable baseline characteristics of the patient groups limit the generalizability of the findings. Larger and well-designed studies are warranted to definitively establish whether dosing strategies for these antibiotics differ between CF and non-CF populations.

4.5. CFTR Modulators

CFTR modulators have been shown to reduce Pa load in the sputum of CF patients [177,178]. These modulators include potentiators, such as Ivacaftor, which maintain the CFTR channels in an open state, allowing chloride ions to flow across cell membranes, and correctors, like Lumacaftor, which help ensure the protein folds correctly and reaches the cell surface [179,180,181]. Although they do not act as antibiotics, restoring CFTR function improves the lung environment, enhancing the effectiveness of antibiotic treatments by improving their penetration into biofilms and reducing the risk of reinfection by facilitating mucus clearance and decreasing bacterial colonization [177]. However, research in adults has shown that while Pa load decreases, chronic infections often persist and remain difficult to fully eradicate, particularly in individuals with advanced lung disease [178].

5. Challenges and Limitations in Treatments

Pa infections remain a major challenge in the management of CF patients, complicating efforts to improve the outcomes in this incurable condition. The genetic and phenotypic diversity of Pa populations in pwCF plays a critical role in treatment challenges and failures. This diversity allows bacterial subpopulations to transition between acute and chronic infection states, driven not only by mutations and the transfer of mobile genetic elements (MGEs) but also by the bacterium’s ability to modulate gene expression in response to environmental cues within the CF lung [182,183,184,185]. This on/off genetic regulation enables Pa to adapt dynamically to fluctuating conditions such as oxygen levels, nutrient availability, and immune pressures. Such adaptability promotes biofilm formation, antibiotic resistance, and immune evasion, allowing the pathogen to persist and thrive in the hostile lung environment. These adaptive strategies are particularly problematic in the context of the underlying pathophysiological conditions in CF patients, where thickened mucus and compromised immune responses create an ideal environment for Pa colonization [186,187,188]. The combined effects of genetic variability, biofilm development, and phenotypic flexibility further complicate efforts to eradicate the bacterium, often leading to persistent and recurrent infections despite aggressive therapy. This underscores the urgent need for continuous monitoring, personalized treatment strategies, and the reassessment of current therapeutic approaches to ensure effective long-term management of Pa infections in CF care [23,189,190,191,192].
Pa can readily develop resistance to most antibiotics. This situation is further complicated by the fact that prolong antibiotic exposure in pwCF selects for aggressive resistant strains, which makes it increasingly difficult to find effective antibiotics for treatment [193]. Pa is also notorious for its ability to form complex structures called biofilms. These complex structures can significantly impede therapeutic efforts by creating a formidable physical barrier around bacterial cell, effectively evading both immune defences and antibiotic action. Bacteria residing within biofilms also exhibit a downregulated metabolic state, leading to a restricted influx of external substrates [194,195]. It has been found that persister cells within the biofilm can tolerate up to 1000× minimum inhibitory concentration (MIC) of antibiotics under in vitro conditions [27,196].
The complex interplay between abnormal mucus production, impaired mucociliary clearance, and biofilm formation by Pa creates a fertile ground for chronic and recalcitrant infections in CF patients which can trigger the development of severe symptoms [197]. The presence of Pa infection is associated with more than 2.5-fold increase in the risk of mortality in CF patients over an 8-year period [198,199]. Acute PEx represents the worsening of symptoms and most common cause of death in CF patients. In fact, the increased frequency of PEx contributes to accelerated lung function decline and diminished quality of life [200,201]. As shown by two independent prognostic models, the higher frequency of annual cystic fibrosis PEx was directly linked to decreased 2-year and 5-year survival rates [202,203]. However, the prompt detection of bacterial cases accompanied by multifaceted management are crucial to minimize the risk, severity, and duration of exacerbations [204].

6. Emerging Treatments

Beyond conventional options, promising emerging therapies are demonstrating potential in the fight against P. aeruginosa infections in CF. Phage therapy is considered to be the most promising approach to treat CF infection [205]. Phages (or bacteriophages) are bacterial viruses with a remarkable degree of host specificity, exclusively targeting and lysing bacterial cells without affecting human cells. They were used as a therapeutic option in the early 20th century, particularly in Eastern Europe, before the widespread discovery and adoption of antibiotics. However, since the challenge of antimicrobial resistance intensifies, phage therapy re-emerges as a promising strategy for managing infections caused by MDR bacteria [206,207].
Gene-editing methods offer significant potential for addressing microbial infections in pwCF, either by fixing the CFTR gene mutation, which is the root cause of complications, or by directly targeting the causative pathogens [208,209,210,211]. CRISPR-Cas technology has emerged as a leading tool in this area. It can be customized to specifically target a CFTR mutation using a guide RNA (gRNA), which directs the Cas enzyme to the defective DNA sequence, enabling correction of the CFTR mutation. This restoration of normal CFTR function improves mucus clearance and reduces the likelihood of chronic infections [209,210]. Furthermore, CRISPR-Cas can be adapted to directly fight Pa infections by targeting bacterial DNA. This can be achieved by designing gRNAs that bind to specific genes responsible for bacterial virulence or antibiotic resistance, leading to the disruption of these genetic elements. As a result, the bacteria become less able to form biofilms or resist antibiotics, making it easier for the immune system to clear the infection [208,211].
Additionally, the development of biofilm-disrupting agents offers a potential solution against chronic bacterial infections [212]. These agents target the structural integrity of the biofilm matrix, which is composed of extracellular polymeric substances (EPS) that shield bacteria from antibiotics and the immune system. These agents can degrade the EPS, inhibit its formation, or disrupt quorum sensing that regulate biofilm development. By breaking down this protective barrier, the bacterial cells become more susceptible to antibiotic treatment and immune clearance [213]. One recent example is the human hormone hANP which has demonstrated promise in dismantling Pa biofilms and enhancing the efficacy of conventional antibiotics [214]. However, large-scale clinical trials are necessary to validate the efficacy and safety of these emerging therapies, especially in the context of CF. Additionally, addressing the potential for phage resistance and ensuring cost-effectiveness of these novel approaches requires further investigation.

7. Conclusions

P. aeruginosa infections continue to pose a significant challenge in CF management. While current antibiotic therapies form the foundation of treatment, their limitations necessitate the exploration of alternative and complementary strategies. Phase IV trials play a crucial role in optimizing current regimens, and promising options like phage therapy and gene editing offer hope for the future. By adopting a multifaceted approach that incorporates these advancements alongside conventional methods, we can achieve an effective control for Pa infection and improve the quality of life for CF patients.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank the Deanship of Scientific Research at Shaqra University for supporting this work.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Quinton, P.M. Cystic fibrosis: Impaired bicarbonate secretion and mucoviscidosis. Lancet 2008, 372, 415–417. [Google Scholar] [CrossRef] [PubMed]
  2. Elborn, J.S. Cystic fibrosis. Lancet 2016, 388, 2519–2531. [Google Scholar] [CrossRef] [PubMed]
  3. Stephenson, A.L.; Stanojevic, S.; Sykes, J.; Burgel, P.-R. The changing epidemiology and demography of cystic fibrosis. Presse Med. 2017, 46, e87–e95. [Google Scholar] [CrossRef]
  4. Guo, J.; Garratt, A.; Hill, A. Worldwide rates of diagnosis and effective treatment for cystic fibrosis. J. Cyst. Fibros. 2022, 21, 456–462. [Google Scholar] [CrossRef]
  5. Castellani, C.; Massie, J.; Sontag, M.; Southern, K.W. Newborn screening for cystic fibrosis. Lancet Respir. Med. 2016, 4, 653–661. [Google Scholar] [CrossRef]
  6. Shteinberg, M.; Haq, I.J.; Polineni, D.; Davies, J.C. Cystic fibrosis. Lancet 2021, 397, 2195–2211. [Google Scholar] [CrossRef]
  7. Ratjen, F.; Bell, S.C.; Rowe, S.M.; Goss, C.H.; Quittner, A.L.; Bush, A. Cystic fibrosis. Nat. Rev. Dis. Primers 2015, 1, 15010–15044. [Google Scholar] [CrossRef]
  8. Grasemann, H.; Ratjen, F. Cystic Fibrosis. N. Engl. J. Med. 2023, 389, 1693–1707. [Google Scholar] [CrossRef]
  9. Sharma, N.; Cutting, G.R. The genetics and genomics of cystic fibrosis. J. Cyst. Fibros. 2020, 19 (Suppl. S1), S5–S9. [Google Scholar] [CrossRef]
  10. Grody, W.W.; Desnick, R.J. Cystic fibrosis population carrier screening: Here at last–are we ready? Genet. Med. 2001, 3, 87–90. [Google Scholar] [CrossRef]
  11. Ioannou, L.; McClaren, B.J.; Massie, J.; Lewis, S.; Metcalfe, S.A.; Forrest, L.; Delatycki, M.B. Population-based carrier screening for cystic fibrosis: A systematic review of 23 years of research. Genet. Med. 2014, 16, 207–216. [Google Scholar] [CrossRef] [PubMed]
  12. Miller, M.E.; Allen, V.M.; Brock, J.-A.K. Incidence and Carrier Frequency of CFTR Gene Mutations in Pregnancies with Echogenic Bowel in Nova Scotia and Prince Edward Island. J. Obstet. Gynaecol. Can. 2018, 40, 896–902. [Google Scholar] [CrossRef] [PubMed]
  13. Kharrazi, M.; Yang, J.; Bishop, T.; Lessing, S.; Young, S.; Graham, S.; Pearl, M.; Chow, H.; Ho, T.; Currier, R.; et al. California Cystic Fibrosis Newborn Screening Consortium Newborn Screening for Cystic Fibrosis in California. Pediatrics 2015, 136, 1062–1072. [Google Scholar] [CrossRef] [PubMed]
  14. Ogden, H.L.; Kim, H.; Wikenheiser-Brokamp, K.A.; Naren, A.P.; Mun, K.S. Cystic Fibrosis Human Organs-on-a-Chip. Micromachines 2021, 12, 747. [Google Scholar] [CrossRef]
  15. Hanssens, L.S.; Duchateau, J.; Casimir, G.J. CFTR Protein: Not Just a Chloride Channel? Cells 2021, 10, 2844. [Google Scholar] [CrossRef]
  16. Turcios, N.L. Cystic Fibrosis Lung Disease: An Overview. Respir. Care 2020, 65, 233–251. [Google Scholar] [CrossRef]
  17. Bergeron, C.; Cantin, A.M. Cystic Fibrosis: Pathophysiology of Lung Disease. Semin. Respir. Crit. Care Med. 2019, 40, 715–726. [Google Scholar] [CrossRef]
  18. Cantin, A.M.; Hartl, D.; Konstan, M.W.; Chmiel, J.F. Inflammation in cystic fibrosis lung disease: Pathogenesis and therapy. J. Cyst. Fibros. 2015, 14, 419–430. [Google Scholar] [CrossRef]
  19. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  20. Bhagirath, A.Y.; Li, Y.; Somayajula, D.; Dadashi, M.; Badr, S.; Duan, K. Cystic fibrosis lung environment and Pseudomonas aeruginosa infection. BMC Pulm. Med. 2016, 16, 174. [Google Scholar] [CrossRef]
  21. Rumpf, C.; Lange, J.; Schwartbeck, B.; Kahl, B.C. Staphylococcus aureus and Cystic Fibrosis-A Close Relationship. What Can We Learn from Sequencing Studies? Pathogens 2021, 10, 1177. [Google Scholar] [CrossRef] [PubMed]
  22. Acosta, N.; Waddell, B.; Heirali, A.; Somayaji, R.; Surette, M.G.; Workentine, M.L.; Rabin, H.R.; Parkins, M.D. Cystic Fibrosis Patients Infected with Epidemic Pseudomonas aeruginosa Strains Have Unique Microbial Communities. Front. Cell. Inf. Microbio. 2020, 10, 173. [Google Scholar] [CrossRef] [PubMed]
  23. Schelstraete, P.; Haerynck, F.; Van daele, S.; Deseyne, S.; De Baets, F. Eradication therapy for Pseudomonas aeruginosa colonization episodes in cystic fibrosis patients not chronically colonized by P. aeruginosa. J. Cyst. Fibros. 2013, 12, 1–8. [Google Scholar] [CrossRef] [PubMed]
  24. Crull, M.R.; Somayaji, R.; Ramos, K.J.; Caldwell, E.; Mayer-Hamblett, N.; Aitken, M.L.; Nichols, D.P.; Rowhani-Rahbar, A.; Goss, C.H. Changing Rates of Chronic Pseudomonas aeruginosa Infections in Cystic Fibrosis: A Population-Based Cohort Study. Clin. Infect. Dis. 2018, 67, 1089–1095. [Google Scholar] [CrossRef] [PubMed]
  25. Ramsey, D.M.; Wozniak, D.J. Understanding the control of Pseudomonas aeruginosa alginate synthesis and the prospects for management of chronic infections in cystic fibrosis. Mol. Microbiol. 2005, 56, 309–322. [Google Scholar] [CrossRef]
  26. Mahajan, S.; Sunsunwal, S.; Gautam, V.; Singh, M.; Ramya, T.N.C. Biofilm inhibitory effect of alginate lyases on mucoid P. aeruginosa from a cystic fibrosis patient. Biochem. Biophys. Rep. 2021, 26, 101028. [Google Scholar] [CrossRef]
  27. Keren, I.; Kaldalu, N.; Spoering, A.; Wang, Y.; Lewis, K. Persister cells and tolerance to antimicrobials. FEMS Microbiol. Lett. 2004, 230, 13–18. [Google Scholar] [CrossRef]
  28. Taccetti, G.; Sly, P.D. Early detection of infection with Pseudomonas aeruginosa in cystic fibrosis: The Holy Grail or an achievable goal? J. Cyst. Fibros. 2014, 13, 491–493. [Google Scholar] [CrossRef]
  29. Douglas, T.A.; Brennan, S.; Gard, S.; Berry, L.; Gangell, C.; Stick, S.M.; Clements, B.S.; Sly, P.D. Acquisition and eradication of P. aeruginosa in young children with cystic fibrosis. Eur. Respir. J. 2009, 33, 305–311. [Google Scholar] [CrossRef]
  30. Jackson, L.; Waters, V. Factors influencing the acquisition and eradication of early Pseudomonas aeruginosa infection in cystic fibrosis. J. Cyst. Fibros. 2021, 20, 8–16. [Google Scholar] [CrossRef]
  31. Kerem, E.; Corey, M.; Gold, R.; Levison, H. Pulmonary function and clinical course in patients with cystic fibrosis after pulmonary colonization with Pseudomonas aeruginosa. J. Pediatr. 1990, 116, 714–719. [Google Scholar] [CrossRef]
  32. Malhotra, S.; Hayes, D.; Wozniak, D.J. Cystic Fibrosis and Pseudomonas aeruginosa: The Host-Microbe Interface. Clin. Microbiol. Rev. 2019, 32, 1–46. [Google Scholar] [CrossRef] [PubMed]
  33. Mésinèle, J.; Ruffin, M.; Kemgang, A.; Guillot, L.; Boëlle, P.-Y.; Corvol, H. Risk factors for Pseudomonas aeruginosa airway infection and lung function decline in children with cystic fibrosis. J. Cyst. Fibros. 2022, 21, 45–51. [Google Scholar] [CrossRef] [PubMed]
  34. Zemanick, E.T.; Bell, S.C. Prevention of chronic infection with Pseudomonas aeruginosa infection in cystic fibrosis. Curr. Opin. Pulm. Med. 2019, 25, 636–645. [Google Scholar] [CrossRef]
  35. Antoniu, S.A. Investigational inhaled therapies for non-CF bronchiectasis. Expert. Opin. Investig. Drugs 2018, 27, 139–146. [Google Scholar] [CrossRef]
  36. Burgener, E.B.; Moss, R.B. Cystic fibrosis transmembrane conductance regulator modulators: Precision medicine in cystic fibrosis. Curr. Opin. Pediatr. 2018, 30, 372–377. [Google Scholar] [CrossRef]
  37. Singh, H.; Jani, C.; Marshall, D.C.; Franco, R.; Bhatt, P.; Podder, S.; Shalhoub, J.; Kurman, J.S.; Nanchal, R.; Uluer, A.Z.; et al. Cystic fibrosis-related mortality in the United States from 1999 to 2020: An observational analysis of time trends and disparities. Sci. Rep. 2023, 13, 15030. [Google Scholar] [CrossRef]
  38. Glasser, S.P.; Salas, M.; Delzell, E. Importance and challenges of studying marketed drugs: What is a phase IV study? Common clinical research designs, registries, and self-reporting systems. J. Clin. Pharmacol. 2007, 47, 1074–1086. [Google Scholar] [CrossRef]
  39. Suvarna, V. Phase IV of Drug Development. Perspect. Clin. Res. 2010, 1, 57–60. [Google Scholar] [CrossRef]
  40. Minneci, P.C.; Deans, K.J. Clinical trials. Semin. Pediatr. Surg. 2018, 27, 332–337. [Google Scholar] [CrossRef]
  41. Southern, K.W.; Addy, C.; Bell, S.C.; Bevan, A.; Borawska, U.; Brown, C.; Burgel, P.-R.; Button, B.; Castellani, C.; Chansard, A.; et al. Standards for the care of people with cystic fibrosis; establishing and maintaining health. J. Cyst. Fibros. 2024, 23, 12–28. [Google Scholar] [CrossRef] [PubMed]
  42. Allen, L.; Allen, L.; Carr, S.B.; Davies, G.; Downey, D.; Egan, M.; Forton, J.T.; Gray, R.; Haworth, C.; Horsley, A.; et al. Future therapies for cystic fibrosis. Nat. Commun. 2023, 14, 693. [Google Scholar] [CrossRef]
  43. Imperlini, E.; Papa, R. Clinical Advances in Cystic Fibrosis. J. Clin. Med. 2022, 11, 6306. [Google Scholar] [CrossRef]
  44. Dienstag, J.; Neu, H.C. In vitro studies of tobramycin, an aminoglycoside antibiotic. Antimicrob. Agents Chemother. 1972, 1, 41–45. [Google Scholar] [CrossRef]
  45. Rosalia, M.; Chiesa, E.; Tottoli, E.M.; Dorati, R.; Genta, I.; Conti, B.; Pisani, S. Tobramycin Nanoantibiotics and Their Advantages: A Minireview. Int. J. Mol. Sci. 2022, 23, 14080. [Google Scholar] [CrossRef]
  46. Kotra, L.P.; Haddad, J.; Mobashery, S. Aminoglycosides: Perspectives on mechanisms of action and resistance and strategies to counter resistance. Antimicrob. Agents Chemother. 2000, 44, 3249–3256. [Google Scholar] [CrossRef]
  47. Food and Drug Administration. TOPI® (Tobramycin Inhalation Solution), for Oral Inhalation Use Initial U.S. Approval: 1975. 2024; pp. 1–13. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/050753s022lbl.pdf (accessed on 10 October 2024).
  48. McDonald, E.M.; Ram, F.S.F.; Patel, D.V.; McGhee, C.N.J. Topical antibiotics for the management of bacterial keratitis: An evidence-based review of high quality randomised controlled trials. Br. J. Ophthalmol. 2014, 98, 1470–1477. [Google Scholar] [CrossRef]
  49. Barker, A.F.; Couch, L.; Fiel, S.B.; Gotfried, M.H.; Ilowite, J.; Meyer, K.C.; O’Donnell, A.; Sahn, S.A.; Smith, L.J.; Stewart, J.O.; et al. Tobramycin solution for inhalation reduces sputum Pseudomonas aeruginosa density in bronchiectasis. Am. J. Respir. Crit. Care Med. 2000, 162, 481–485. [Google Scholar] [CrossRef]
  50. Lam, J.; Vaughan, S.; Parkins, M.D. Tobramycin Inhalation Powder (TIP): An Efficient Treatment Strategy for the Management of Chronic Pseudomonas aeruginosa Infection in Cystic Fibrosis. Clin. Med. Insights Circ. Respir. Pulm. Med. 2013, 7, 61–77. [Google Scholar] [CrossRef]
  51. Banerjee, S.K.; Jagannath, C.; Hunter, R.L.; Dasgupta, A. Bioavailability of tobramycin after oral delivery in FVB mice using CRL-1605 copolymer, an inhibitor of P-glycoprotein. Life Sci. 2000, 67, 2011–2016. [Google Scholar] [CrossRef]
  52. Hennig, S.; McKay, K.; Vidmar, S.; O’Brien, K.; Stacey, S.; Cheney, J.; Wainwright, C.E. Safety of inhaled (Tobi®) and intravenous tobramycin in young children with cystic fibrosis. J. Cyst. Fibros. 2014, 13, 428–434. [Google Scholar] [CrossRef] [PubMed]
  53. Smith, A.L.; Ramsey, B.W.; Hedges, D.L.; Hack, B.; Williams-Warren, J.; Weber, A.; Gore, E.J.; Redding, G.J. Safety of aerosol tobramycin administration for 3 months to patients with cystic fibrosis. Pediatr. Pulmonol. 1989, 7, 265–271. [Google Scholar] [CrossRef]
  54. Ramsey, B.W.; Dorkin, H.L.; Eisenberg, J.D.; Gibson, R.L.; Harwood, I.R.; Kravitz, R.M.; Schidlow, D.V.; Wilmott, R.W.; Astley, S.J.; McBurnie, M.A. Efficacy of aerosolized tobramycin in patients with cystic fibrosis. N. Engl. J. Med. 1993, 328, 1740–1746. [Google Scholar] [CrossRef]
  55. Shteinberg, M.; Elborn, J.S. Use of inhaled tobramycin in cystic fibrosis. Adv. Ther. 2015, 32, 1–9. [Google Scholar] [CrossRef]
  56. Bowman, C.M. The long-term use of inhaled tobramycin in patients with cystic fibrosis. J. Cyst. Fibros. 2002, 1, 194–198. [Google Scholar] [CrossRef]
  57. Schwarz, C.; Taccetti, G.; Burgel, P.-R.; Mulrennan, S. Tobramycin safety and efficacy review article. Respir. Med. 2022, 195, 106778. [Google Scholar] [CrossRef]
  58. Bialvaei, A.Z.; Samadi Kafil, H. Colistin, mechanisms and prevalence of resistance. Curr. Med. Res. Opin. 2015, 31, 707–721. [Google Scholar] [CrossRef]
  59. Ito, M.; Aida, K.; Uemura, T. Biosynthesis of colistin by Bacillus colistinus Koyama. Biochim. Biophys. Acta 1970, 213, 244–247. [Google Scholar] [CrossRef]
  60. Bertani, B.; Ruiz, N. Function and Biogenesis of Lipopolysaccharides. EcoSal Plus 2018, 8, 1–33. [Google Scholar] [CrossRef]
  61. Liao, F.-H.; Wu, T.-H.; Yao, C.-N.; Kuo, S.-C.; Su, C.-J.; Jeng, U.-S.; Lin, S.-Y. A Supramolecular Trap to Increase the Antibacterial Activity of Colistin. Angew. Chem. Int. Ed. Engl. 2020, 59, 1430–1434. [Google Scholar] [CrossRef]
  62. El-Sayed Ahmed, M.A.E.-G.; Zhong, L.-L.; Shen, C.; Yang, Y.; Doi, Y.; Tian, G.-B. Colistin and its role in the Era of antibiotic resistance: An extended review (2000–2019). Emerg. Microbes Infect. 2020, 9, 868–885. [Google Scholar] [CrossRef] [PubMed]
  63. Jeannot, K.; Bolard, A.; Plésiat, P. Resistance to polymyxins in Gram-negative organisms. Int. J. Antimicrob. Agents 2017, 49, 526–535. [Google Scholar] [CrossRef] [PubMed]
  64. Srinivas, P.; Rivard, K. Polymyxin Resistance in Gram-negative Pathogens. Curr. Infect. Dis. Rep. 2017, 19, 38–39. [Google Scholar] [CrossRef]
  65. Dudhani, R.V.; Nation, R.L.; Li, J. Evaluating the stability of colistin and colistin methanesulphonate in human plasma under different conditions of storage. J. Antimicrob. Chemother. 2010, 65, 1412–1415. [Google Scholar] [CrossRef] [PubMed]
  66. Bergen, P.J.; Li, J.; Rayner, C.R.; Nation, R.L. Colistin methanesulfonate is an inactive prodrug of colistin against Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2006, 50, 1953–1958. [Google Scholar] [CrossRef] [PubMed]
  67. Tewes, F.; Brillault, J.; Gregoire, N.; Olivier, J.-C.; Lamarche, I.; Adier, C.; Healy, A.-M.; Marchand, S. Comparison between Colistin Sulfate Dry Powder and Solution for Pulmonary Delivery. Pharmaceutics 2020, 12, 557. [Google Scholar] [CrossRef]
  68. Spapen, H.; Jacobs, R.; Van Gorp, V.; Troubleyn, J.; Honoré, P.M. Renal and neurological side effects of colistin in critically ill patients. Ann. Intensive Care 2011, 1, 14–17. [Google Scholar] [CrossRef]
  69. Wiseman, L.R.; Wagstaff, A.J.; Brogden, R.N.; Bryson, H.M. Meropenem. A review of its antibacterial activity, pharmacokinetic properties and clinical efficacy. Drugs 1995, 50, 73–101. [Google Scholar] [CrossRef]
  70. Aurilio, C.; Sansone, P.; Barbarisi, M.; Pota, V.; Giaccari, L.G.; Coppolino, F.; Barbarisi, A.; Passavanti, M.B.; Pace, M.C. Mechanisms of Action of Carbapenem Resistance. Antibiotics 2022, 11, 421. [Google Scholar] [CrossRef]
  71. Dhillon, S. Meropenem/Vaborbactam: A Review in Complicated Urinary Tract Infections. Drugs 2018, 78, 1259–1270. [Google Scholar] [CrossRef]
  72. Sanders, C.C.; Sanders, W.E.; Thomson, K.S.; Iaconis, J.P. Meropenem: Activity against resistant gram-negative bacteria and interactions with beta-lactamases. J. Antimicrob. Chemother. 1989, 24 (Suppl. A), 187–196. [Google Scholar] [CrossRef]
  73. Linden, P. Safety profile of meropenem: An updated review of over 6,000 patients treated with meropenem. Drug Saf. 2007, 30, 657–668. [Google Scholar] [CrossRef] [PubMed]
  74. Raza, A.; Ngieng, S.C.; Sime, F.B.; Cabot, P.J.; Roberts, J.A.; Popat, A.; Kumeria, T.; Falconer, J.R. Oral meropenem for superbugs: Challenges and opportunities. Drug Discov. Today 2021, 26, 551–560. [Google Scholar] [CrossRef] [PubMed]
  75. Norrby, S.R.; Gildon, K.M. Safety profile of meropenem: A review of nearly 5,000 patients treated with meropenem. Scand. J. Infect. Dis. 1999, 31, 3–10. [Google Scholar] [PubMed]
  76. Bachar, S.C.; Pal, T. Estimation of Meropenem in Human Plasma by HPLC-UV and its Application in Comparative Bioavailability Study. J. Anal. Chem. 2011, 1, 23–30. [Google Scholar]
  77. Bergan, T.; Nord, C.E.; Thorsteinsson, S.B. Effect of meropenem on the intestinal microflora. Eur. J. Clin. Microbiol. Infect. Dis. 1991, 10, 524–527. [Google Scholar] [CrossRef]
  78. Rafey, A.; Jahan, S.; Farooq, U.; Akhtar, F.; Irshad, M.; Nizamuddin, S.; Parveen, A. Antibiotics Associated with Clostridium difficile Infection. Cureus 2023, 15, e39029. [Google Scholar] [CrossRef]
  79. López Montesinos, I.; Montero, M.; Sorlí, L.; Horcajada, J.P. Ceftolozane-tazobactam: When, how and why using it? Rev. Esp. Quim. 2021, 34 (Suppl. S1), 35–37. [Google Scholar] [CrossRef]
  80. Lizza, B.D.; Betthauser, K.D.; Ritchie, D.J.; Micek, S.T.; Kollef, M.H. New Perspectives on Antimicrobial Agents: Ceftolozane-Tazobactam. Antimicrob. Agents Chemother. 2021, 65, e0231820. [Google Scholar] [CrossRef]
  81. Food and Drug Administration. ZERBAXA® (Ceftolozane and Tazobactam) for Injection, for Intravenous Use. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/206829s011s012lbl.pdf (accessed on 10 October 2024).
  82. European Medicines Agency. Zerbaxa, INN-Ceftolozane/Tazobactam. Available online: https://www.ema.europa.eu/en/documents/product-information/zerbaxa-epar-product-information_en.pdf (accessed on 10 October 2024).
  83. Solomkin, J.; Hershberger, E.; Miller, B.; Popejoy, M.; Friedland, I.; Steenbergen, J.; Yoon, M.; Collins, S.; Yuan, G.; Barie, P.S.; et al. Ceftolozane/Tazobactam Plus Metronidazole for Complicated Intra-abdominal Infections in an Era of Multidrug Resistance: Results from a Randomized, Double-Blind, Phase 3 Trial (ASPECT-cIAI). Clin. Infect. Dis. 2015, 60, 1462–1471. [Google Scholar] [CrossRef]
  84. Bakheit, A.H.H.; Al-Hadiya, B.M.H.; Abd-Elgalil, A.A. Azithromycin, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2014; Volume 39, pp. 1–40. [Google Scholar]
  85. Steingrimsson, O.; Olafsson, J.H.; Thorarinsson, H.; Ryan, R.W.; Johnson, R.B.; Tilton, R.C. Azithromycin in the treatment of sexually transmitted disease. J. Antimicrob. Chemother. 1990, 25 (Suppl. A), 109–114. [Google Scholar] [CrossRef] [PubMed]
  86. Parnham, M.J.; Erakovic Haber, V.; Giamarellos-Bourboulis, E.J.; Perletti, G.; Verleden, G.M.; Vos, R. Azithromycin: Mechanisms of action and their relevance for clinical applications. Pharmacol. Ther. 2014, 143, 225–245. [Google Scholar] [CrossRef] [PubMed]
  87. Goldstein, F.W.; Emirian, M.F.; Coutrot, A.; Acar, J.F. Bacteriostatic and bactericidal activity of azithromycin against Haemophilus influenzae. J. Antimicrob. Chemother. 1990, 25 (Suppl. A), 25–28. [Google Scholar] [CrossRef] [PubMed]
  88. Neu, H.C. Clinical microbiology of azithromycin. Am. J. Med. 1991, 91, 12S–18S. [Google Scholar] [CrossRef]
  89. Sun, J.; Li, Y. Long-term, low-dose macrolide antibiotic treatment in pediatric chronic airway diseases. Pediatr. Res. 2022, 91, 1036–1042. [Google Scholar] [CrossRef]
  90. Cramer, C.L.; Patterson, A.; Alchakaki, A.; Soubani, A.O. Immunomodulatory indications of azithromycin in respiratory disease: A concise review for the clinician. Postgrad. Med. 2017, 129, 493–499. [Google Scholar] [CrossRef]
  91. Stellari, F.F.; Sala, A.; Donofrio, G.; Ruscitti, F.; Caruso, P.; Topini, T.M.; Francis, K.P.; Li, X.; Carnini, C.; Civelli, M.; et al. Azithromycin inhibits nuclear factor-κB activation during lung inflammation: An in vivo imaging study. Pharmacol. Res. Perspect. 2014, 2, e00058. [Google Scholar] [CrossRef]
  92. Aghai, Z.H.; Kode, A.; Saslow, J.G.; Nakhla, T.; Farhath, S.; Stahl, G.E.; Eydelman, R.; Strande, L.; Leone, P.; Rahman, I. Azithromycin suppresses activation of nuclear factor-kappa B and synthesis of pro-inflammatory cytokines in tracheal aspirate cells from premature infants. Pediatr. Res. 2007, 62, 483–488. [Google Scholar] [CrossRef]
  93. Murphy, B.S.; Sundareshan, V.; Cory, T.J.; Hayes, D.; Anstead, M.I.; Feola, D.J. Azithromycin alters macrophage phenotype. J. Antimicrob. Chemother. 2008, 61, 554–560. [Google Scholar] [CrossRef]
  94. Feola, D.J.; Garvy, B.A.; Cory, T.J.; Birket, S.E.; Hoy, H.; Hayes, D.; Murphy, B.S. Azithromycin alters macrophage phenotype and pulmonary compartmentalization during lung infection with Pseudomonas. Antimicrob. Agents Chemother. 2010, 54, 2437–2447. [Google Scholar] [CrossRef]
  95. Bakar, O.; Demirçay, Z.; Yuksel, M.; Haklar, G.; Sanisoglu, Y. The effect of azithromycin on reactive oxygen species in rosacea. Clin. Exp. Dermatol. 2007, 32, 197–200. [Google Scholar] [CrossRef] [PubMed]
  96. Pollock, J.; Chalmers, J.D. The immunomodulatory effects of macrolide antibiotics in respiratory disease. Pulm. Pharmacol. Ther. 2021, 71, 102095. [Google Scholar] [CrossRef] [PubMed]
  97. Paris, R.; Confalonieri, M.; Dal Negro, R.; Ligia, G.P.; Mos, L.; Todisco, T.; Rastelli, V.; Perna, G.; Cepparulo, M. Efficacy and safety of azithromycin 1 g once daily for 3 days in the treatment of community-acquired pneumonia: An open-label randomised comparison with amoxicillin-clavulanate 875/125 mg twice daily for 7 days. J. Chemother. 2008, 20, 77–86. [Google Scholar] [CrossRef]
  98. Garey, K.W.; Amsden, G.W. Intravenous azithromycin. Ann. Pharmacother. 1999, 33, 218–228. [Google Scholar] [CrossRef]
  99. Amsden, G.W. Erythromycin, clarithromycin, and azithromycin: Are the differences real? Clin. Ther. 1996, 18, 56–72, discussion 55. [Google Scholar] [CrossRef]
  100. Hopkins, S. Clinical toleration and safety of azithromycin. Am. J. Med. 1991, 91, 40S–45S. [Google Scholar] [CrossRef]
  101. Sun, Y.-W.; Cen, Y.-H.; Chen, M.-H.; Yan, X.-K.; Jin, X.-F. Safety profiles and adverse reactions of azithromycin in the treatment of pediatric respiratory diseases: A systematic review and meta-analysis. Medicine 2023, 102, e36306. [Google Scholar] [CrossRef]
  102. Zhanel, G.G.; Walkty, A.; Vercaigne, L.; Karlowsky, J.A.; Embil, J.; Gin, A.S.; Hoban, D.J. The new fluoroquinolones: A critical review. Can. J. Infect. Dis. 1999, 10, 207–238. [Google Scholar] [CrossRef]
  103. King, D.E.; Malone, R.; Lilley, S.H. New classification and update on the quinolone antibiotics. Am. Fam. Physician 2000, 61, 2741–2748. [Google Scholar]
  104. Hooper, D.C.; Jacoby, G.A. Topoisomerase Inhibitors: Fluoroquinolone Mechanisms of Action and Resistance. Cold Spring Harb. Perspect. Med. 2016, 6, a025320. [Google Scholar] [CrossRef]
  105. Hooper, D.C. Mechanisms of action and resistance of older and newer fluoroquinolones. Clin. Infect. Dis. 2000, 31 (Suppl. S2), S24–S28. [Google Scholar] [CrossRef] [PubMed]
  106. Yudhawati, R.; Wicaksono, N.F. Immunomodulatory Effects of Fluoroquinolones in Community-Acquired Pneumonia-Associated Acute Respiratory Distress Syndrome. Biomedicines 2024, 12, 761. [Google Scholar] [CrossRef]
  107. Dorkin, H.L.; Staab, D.; Operschall, E.; Alder, J.; Criollo, M. Ciprofloxacin DPI: A randomised, placebo-controlled, phase IIb efficacy and safety study on cystic fibrosis. BMJ Open Respir. Res. 2015, 2, e000100. [Google Scholar] [CrossRef]
  108. Cao, D.; Shen, Y.; Huang, Y.; Chen, B.; Chen, Z.; Ai, J.; Liu, L.; Yang, L.; Wei, Q. Levofloxacin Versus Ciprofloxacin in the Treatment of Urinary Tract Infections: Evidence-Based Analysis. Front. Pharmacol. 2021, 12, 658095. [Google Scholar] [CrossRef]
  109. Tribble, D.R. Antibiotic Therapy for Acute Watery Diarrhea and Dysentery. Mil. Med. 2017, 182, 17–25. [Google Scholar] [CrossRef]
  110. Thai, T.; Salisbury, B.H.; Zito, P.M. Ciprofloxacin; StatPearls Publishing: Treasure Island, FL, USA, 2024; pp. 1–9. [Google Scholar]
  111. Ball, P. Ciprofloxacin: An overview of adverse experiences. J. Antimicrob. Chemother. 1986, 18 (Suppl. D), 187–193. [Google Scholar] [CrossRef]
  112. Agbaht, K.; Bitik, B.; Piskinpasa, S.; Bayraktar, M.; Topeli, A. Ciprofloxacin-associated seizures in a patient with underlying thyrotoxicosis: Case report and literature review. Int. J. Clin. Pharmacol. Ther. 2009, 47, 303–310. [Google Scholar] [CrossRef]
  113. Berger, F.A.; Monadian, N.; de Groot, N.M.S.; Santbergen, B.; van der Sijs, H.; Becker, M.L.; Broers, A.E.C.; van Gelder, T.; van den Bemt, P.M.L.A. QTc prolongation during ciprofloxacin and fluconazole combination therapy: Prevalence and associated risk factors. Br. J. Clin. Pharmacol. 2018, 84, 369–378. [Google Scholar] [CrossRef]
  114. Adefurin, A.; Sammons, H.; Jacqz-Aigrain, E.; Choonara, I. Ciprofloxacin safety in paediatrics: A systematic review. Arch. Dis. Child. 2011, 96, 874–880. [Google Scholar] [CrossRef]
  115. Yefet, E.; Schwartz, N.; Chazan, B.; Salim, R.; Romano, S.; Nachum, Z. The safety of quinolones and fluoroquinolones in pregnancy: A meta-analysis. BJOG 2018, 125, 1069–1076. [Google Scholar] [CrossRef]
  116. Ramsey, C.; MacGowan, A.P. A review of the pharmacokinetics and pharmacodynamics of aztreonam. J. Antimicrob. Chemother. 2016, 71, 2704–2712. [Google Scholar] [CrossRef] [PubMed]
  117. Stutman, H.R. Clinical experience with aztreonam. Pediatr. Infect. Dis. J. 1989, 8, S109–S112, discussion S128–S132. [Google Scholar] [CrossRef] [PubMed]
  118. Paul, M.; Carrara, E.; Retamar, P.; Tängdén, T.; Bitterman, R.; Bonomo, R.A.; de Waele, J.; Daikos, G.L.; Akova, M.; Harbarth, S.; et al. European Society of Clinical Microbiology and Infectious Diseases (ESCMID) guidelines for the treatment of infections caused by multidrug-resistant Gram-negative bacilli (endorsed by European society of intensive care medicine). Clin. Microbiol. Infect. 2022, 28, 521–547. [Google Scholar] [CrossRef]
  119. Crichton, M.L.; Lonergan, M.; Barker, A.F.; Sibila, O.; Goeminne, P.; Shoemark, A.; Chalmers, J.D. Inhaled aztreonam improves symptoms of cough and sputum production in patients with bronchiectasis: A post hocanalysis of the AIR-BX studies. Eur. Respir. J. 2020, 56, 1–9. [Google Scholar] [CrossRef]
  120. Delp, H.; Gibson, G.A.; Buckman, S.A. Aztreonam-avibactam for the treatment of intra-abdominal infections. Expert. Opin. Pharmacother. 2024, 25, 1867–1872. [Google Scholar] [CrossRef]
  121. Sattler, F.R.; Moyer, J.E.; Schramm, M.; Lombard, J.S.; Appelbaum, P.C. Aztreonam compared with gentamicin for treatment of serious urinary tract infections. Lancet 1984, 1, 1315–1318. [Google Scholar] [CrossRef]
  122. Newman, T.J.; Dreslinski, G.R.; Tadros, S.S. Safety profile of aztreonam in clinical trials. Rev. Infect. Dis. 1985, 7 (Suppl. S4), S648–S655. [Google Scholar] [CrossRef]
  123. Manos, J. Current and Emerging Therapies to Combat Cystic Fibrosis Lung Infections. Microorganisms 2021, 9, 1874. [Google Scholar] [CrossRef]
  124. Graña-Miraglia, L.; Morales-Lizcano, N.; Wang, P.W.; Hwang, D.M.; Yau, Y.C.W.; Waters, V.J.; Guttman, D.S. Predictive modeling of antibiotic eradication therapy success for new-onset Pseudomonas aeruginosa pulmonary infections in children with cystic fibrosis. PLoS Comput. Biol. 2023, 19, e1011424. [Google Scholar] [CrossRef]
  125. Taccetti, G.; Campana, S.; Festini, F.; Mascherini, M.; Döring, G. Early eradication therapy against Pseudomonas aeruginosa in cystic fibrosis patients. Eur. Respir. J. 2005, 26, 458–461. [Google Scholar] [CrossRef]
  126. Jones, A.M. Eradication therapy for early Pseudomonas aeruginosa infection in CF: Many questions still unanswered. Eur. Respir. J. 2005, 26, 373–375. [Google Scholar] [CrossRef] [PubMed]
  127. Akkerman-Nijland, A.M.; Yousofi, M.; Rottier, B.L.; Van der Vaart, H.; Burgerhof, J.G.M.; Frijlink, H.W.; Touw, D.J.; Koppelman, G.H.; Akkerman, O.W. Eradication of Pseudomonas aeruginosa in cystic fibrosis patients with inhalation of dry powder tobramycin. Ther. Adv. Respir. Dis. 2020, 14, 1753466620905279-5. [Google Scholar] [CrossRef] [PubMed]
  128. Ratjen, F.; Munck, A.; Kho, P.; Angyalosi, G.; ELITE Study Group. Treatment of early Pseudomonas aeruginosa infection in patients with cystic fibrosis: The ELITE trial. Thorax 2010, 65, 286–291. [Google Scholar] [CrossRef]
  129. Conceição, M.; Shteinberg, M.; Goeminne, P.; Altenburg, J.; Chalmers, J.D. Eradication treatment for Pseudomonas aeruginosa infection in adults with bronchiectasis: A systematic review and meta-analysis. Eur. Respir. Rev. 2024, 33, 1–9. [Google Scholar] [CrossRef]
  130. McCarthy, K.; Avent, M. Oral or intravenous antibiotics? Aust. Prescr. 2020, 43, 45–48. [Google Scholar] [CrossRef]
  131. Molinelli, E.; De Simoni, E.; Candelora, M.; Sapigni, C.; Brisigotti, V.; Rizzetto, G.; Offidani, A.; Simonetti, O. Systemic Antibiotic Therapy in Hidradenitis Suppurativa: A Review on Treatment Landscape and Current Issues. Antibiotics 2023, 12, 978. [Google Scholar] [CrossRef]
  132. Yuan, X.; Zhou, F.; Wang, H.; Xu, X.; Xu, S.; Zhang, C.; Zhang, Y.; Lu, M.; Zhang, Y.; Zhou, M.; et al. Systemic antibiotics increase microbiota pathogenicity and oral bone loss. Int. J. Oral. Sci. 2023, 15, 4–14. [Google Scholar] [CrossRef]
  133. Gounden, R.; Bamford, C.; van Zyl-Smit, R.; Cohen, K.; Maartens, G. Safety and effectiveness of colistin compared with tobramycin for multi-drug resistant Acinetobacter baumannii infections. BMC Infect. Dis. 2009, 9, 26. [Google Scholar] [CrossRef]
  134. McWilliam, S.J.; Antoine, D.J.; Smyth, R.L.; Pirmohamed, M. Aminoglycoside-induced nephrotoxicity in children. Pediatr. Nephrol. 2017, 32, 2015–2025. [Google Scholar] [CrossRef]
  135. Spencer, S.; Ipema, H.; Hartke, P.; Krueger, C.; Rodriguez, R.; Gross, A.E.; Gabay, M. Intravenous Push Administration of Antibiotics: Literature and Considerations. Hosp. Pharm. 2018, 53, 157–169. [Google Scholar] [CrossRef]
  136. Ordooei Javan, A.; Shokouhi, S.; Sahraei, Z. A review on colistin nephrotoxicity. Eur. J. Clin. Pharmacol. 2015, 71, 801–810. [Google Scholar] [CrossRef] [PubMed]
  137. Glass, S.; Plant, N.D.; Spencer, D.A. The effects of intravenous tobramycin on renal tubular function in children with cystic fibrosis. J. Cyst. Fibros. 2005, 4, 221–225. [Google Scholar] [CrossRef] [PubMed]
  138. Özkarakaş, H.; Köse, I.; Zincircioğlu, Ç.; Ersan, S.; Ersan, G.; Şenoğlu, N.; Köse, Ş.; Erbay, R.H. Risk factors for colistin-associated nephrotoxicity and mortality in critically ill patients. Turk. J. Med. Sci. 2017, 47, 1165–1172. [Google Scholar] [CrossRef] [PubMed]
  139. Nation, R.L.; Li, J. Colistin in the 21st century. Curr. Opin. Infect. Dis. 2009, 22, 535–543. [Google Scholar] [CrossRef]
  140. Falagas, M.E.; Kasiakou, S.K.; Tsiodras, S.; Michalopoulos, A. The use of intravenous and aerosolized polymyxins for the treatment of infections in critically ill patients: A review of the recent literature. Clin. Med. Res. 2006, 4, 138–146. [Google Scholar] [CrossRef]
  141. Hartzell, J.D.; Neff, R.; Ake, J.; Howard, R.; Olson, S.; Paolino, K.; Vishnepolsky, M.; Weintrob, A.; Wortmann, G. Nephrotoxicity associated with intravenous colistin (colistimethate sodium) treatment at a tertiary care medical center. Clin. Infect. Dis. 2009, 48, 1724–1728. [Google Scholar] [CrossRef]
  142. Koomanachai, P.; Tiengrim, S.; Kiratisin, P.; Thamlikitkul, V. Efficacy and safety of colistin (colistimethate sodium) for therapy of infections caused by multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii in Siriraj Hospital, Bangkok, Thailand. Int. J. Infect. Dis. 2007, 11, 402–406. [Google Scholar] [CrossRef]
  143. Minejima, E.; Choi, J.; Beringer, P.; Lou, M.; Tse, E.; Wong-Beringer, A. Applying new diagnostic criteria for acute kidney injury to facilitate early identification of nephrotoxicity in vancomycin-treated patients. Antimicrob. Agents Chemother. 2011, 55, 3278–3283. [Google Scholar] [CrossRef]
  144. Ramsey, B.W.; Pepe, M.S.; Quan, J.M.; Otto, K.L.; Montgomery, A.B.; Williams-Warren, J.; Vasiljev-K, M.; Borowitz, D.; Bowman, C.M.; Marshall, B.C.; et al. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. Cystic Fibrosis Inhaled Tobramycin Study Group. N. Engl. J. Med. 1999, 340, 23–30. [Google Scholar] [CrossRef]
  145. Anderson, S.; Atkins, P.; Bäckman, P.; Cipolla, D.; Clark, A.; Daviskas, E.; Disse, B.; Entcheva-Dimitrov, P.; Fuller, R.; Gonda, I.; et al. Inhaled Medicines: Past, Present, and Future. Pharmacol. Rev. 2022, 74, 48–118. [Google Scholar] [CrossRef]
  146. Sommerwerck, U.; Virella-Lowell, I.; Angyalosi, G.; Viegas, A.; Cao, W.; Debonnett, L. Long-term safety of tobramycin inhalation powder in patients with cystic fibrosis: Phase IV (ETOILES) study. Curr. Med. Res. Opin. 2016, 32, 1789–1795. [Google Scholar] [CrossRef] [PubMed]
  147. Konstan, M.W.; Geller, D.E.; Minić, P.; Brockhaus, F.; Zhang, J.; Angyalosi, G. Tobramycin inhalation powder for P. aeruginosa infection in cystic fibrosis: The EVOLVE trial. Pediatr. Pulmonol. 2011, 46, 230–238. [Google Scholar] [CrossRef] [PubMed]
  148. Konstan, M.W.; Flume, P.A.; Kappler, M.; Chiron, R.; Higgins, M.; Brockhaus, F.; Zhang, J.; Angyalosi, G.; He, E.; Geller, D.E. Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: The EAGER trial. J. Cyst. Fibros. 2011, 10, 54–61. [Google Scholar] [CrossRef] [PubMed]
  149. Chalmers, J.D.; Smith, M.P.; McHugh, B.J.; Doherty, C.; Govan, J.R.; Hill, A.T. Short- and long-term antibiotic treatment reduces airway and systemic inflammation in non-cystic fibrosis bronchiectasis. Am. J. Respir. Crit. Care Med. 2012, 186, 657–665. [Google Scholar] [CrossRef]
  150. Cordeiro, R.; Choi, H.; Haworth, C.S.; Chalmers, J.D. The Efficacy and Safety of Inhaled Antibiotics for the Treatment of Bronchiectasis in Adults: Updated Systematic Review and Meta-Analysis. CHEST 2024, 166, 61–80. [Google Scholar] [CrossRef]
  151. Wenzler, E.; Fraidenburg, D.R.; Scardina, T.; Danziger, L.H. Inhaled Antibiotics for Gram-Negative Respiratory Infections. Clin. Microbiol. Rev. 2016, 29, 581–632. [Google Scholar] [CrossRef]
  152. Thompson, V.; Mayer-Hamblett, N.; Kloster, M.; Bilton, D.; Flume, P.A. Risk of hemoptysis in cystic fibrosis clinical trials: A retrospective cohort study. J. Cyst. Fibros. 2015, 14, 632–638. [Google Scholar] [CrossRef]
  153. Maselli, D.J.; Keyt, H.; Restrepo, M.I. Inhaled Antibiotic Therapy in Chronic Respiratory Diseases. Int. J. Mol. Sci. 2017, 18, 1062. [Google Scholar] [CrossRef]
  154. Greenwood, J.; Schwarz, C.; Sommerwerck, U.; Nash, E.F.; Tamm, M.; Cao, W.; Mastoridis, P.; Debonnett, L.; Hamed, K. Ease of use of tobramycin inhalation powder compared with nebulized tobramycin and colistimethate sodium: A crossover study in cystic fibrosis patients with pulmonary Pseudomonas aeruginosa infection. Ther. Adv. Respir. Dis. 2017, 11, 249–260. [Google Scholar] [CrossRef]
  155. Harrison, M.J.; McCarthy, M.; Fleming, C.; Hickey, C.; Shortt, C.; Eustace, J.A.; Murphy, D.M.; Plant, B.J. Inhaled versus nebulised tobramycin: A real world comparison in adult cystic fibrosis (CF). J. Cyst. Fibros. 2014, 13, 692–698. [Google Scholar] [CrossRef]
  156. von Schantz, S.; Katajavuori, N.; Antikainen, O.; Juppo, A. Evaluation of dry powder inhalers with a focus on ease of use and user preference in inhaler-naïve individuals. Int. J. Pharm. 2016, 509, 50–58. [Google Scholar] [CrossRef] [PubMed]
  157. Schuster, A.; Haliburn, C.; Döring, G.; Goldman, M.H.; Freedom Study Group. Safety, efficacy and convenience of colistimethate sodium dry powder for inhalation (Colobreathe DPI) in patients with cystic fibrosis: A randomised study. Thorax 2013, 68, 344–350. [Google Scholar] [CrossRef] [PubMed]
  158. Galeva, I.; Konstan, M.W.; Higgins, M.; Angyalosi, G.; Brockhaus, F.; Piggott, S.; Thomas, K.; Chuchalin, A.G. Tobramycin inhalation powder manufactured by improved process in cystic fibrosis: The randomized EDIT trial. Curr. Med. Res. Opin. 2013, 29, 947–956. [Google Scholar] [CrossRef] [PubMed]
  159. de la Rosa-Carrillo, D.; Suárez-Cuartín, G.; Sibila, O.; Golpe, R.; Girón, R.-M.; Martinez-García, M.A. Efficacy and Safety of Dry Powder Antibiotics: A Narrative Review. J. Clin. Med. 2023, 12, 3577. [Google Scholar] [CrossRef] [PubMed]
  160. Geller, D.E.; Weers, J.; Heuerding, S. Development of an inhaled dry-powder formulation of tobramycin using PulmoSphere™ technology. J. Aerosol Med. Pulm. Drug Deliv. 2011, 24, 175–182. [Google Scholar] [CrossRef]
  161. Conole, D.; Keating, G.M. Colistimethate sodium dry powder for inhalation: A review of its use in the treatment of chronic Pseudomonas aeruginosa infection in patients with cystic fibrosis. Drugs 2014, 74, 377–387. [Google Scholar] [CrossRef]
  162. Coates, A.R.M.; Hu, Y.; Holt, J.; Yeh, P. Antibiotic combination therapy against resistant bacterial infections: Synergy, rejuvenation and resistance reduction. Expert. Rev. Anti-Infect. Ther. 2020, 18, 5–15. [Google Scholar] [CrossRef]
  163. Nick, J.A.; Moskowitz, S.M.; Chmiel, J.F.; Forssén, A.V.; Kim, S.H.; Saavedra, M.T.; Saiman, L.; Taylor-Cousar, J.L.; Nichols, D.P. Azithromycin may antagonize inhaled tobramycin when targeting Pseudomonas aeruginosa in cystic fibrosis. Ann. Am. Thorac. Soc. 2014, 11, 342–350. [Google Scholar] [CrossRef]
  164. Nichols, D.P.; Singh, P.K.; Baines, A.; Caverly, L.J.; Chmiel, J.F.; Gibson, R.L.; Lascano, J.; Morgan, S.J.; Retsch-Bogart, G.; Saiman, L.; et al. Testing the effects of combining azithromycin with inhaled tobramycin for P. aeruginosa in cystic fibrosis: A randomised, controlled clinical trial. Thorax 2022, 77, 581–588. [Google Scholar] [CrossRef]
  165. Mayer-Hamblett, N.; Retsch-Bogart, G.; Kloster, M.; Accurso, F.; Rosenfeld, M.; Albers, G.; Black, P.; Brown, P.; Cairns, A.; Davis, S.D.; et al. Azithromycin for Early Pseudomonas Infection in Cystic Fibrosis. The OPTIMIZE Randomized Trial. Am. J. Respir. Crit. Care Med. 2018, 198, 1177–1187. [Google Scholar] [CrossRef]
  166. Equi, A.; Balfour-Lynn, I.M.; Bush, A.; Rosenthal, M. Long term azithromycin in children with cystic fibrosis: A randomised, placebo-controlled crossover trial. Lancet 2002, 360, 978–984. [Google Scholar] [CrossRef] [PubMed]
  167. Clement, A.; Tamalet, A.; Leroux, E.; Ravilly, S.; Fauroux, B.; Jais, J.-P. Long term effects of azithromycin in patients with cystic fibrosis: A double blind, placebo controlled trial. Thorax 2006, 61, 895–902. [Google Scholar] [CrossRef] [PubMed]
  168. Stick, S.M.; Foti, A.; Ware, R.S.; Tiddens, H.A.W.M.; Clements, B.S.; Armstrong, D.S.; Selvadurai, H.; Tai, A.; Cooper, P.J.; Byrnes, C.A.; et al. The effect of azithromycin on structural lung disease in infants with cystic fibrosis (COMBAT CF): A phase 3, randomised, double-blind, placebo-controlled clinical trial. Lancet Respir. Med. 2022, 10, 776–784. [Google Scholar] [CrossRef]
  169. Hennig, S.; Standing, J.F.; Staatz, C.E.; Thomson, A.H. Population pharmacokinetics of tobramycin in patients with and without cystic fibrosis. Clin. Pharmacokinet. 2013, 52, 289–301. [Google Scholar] [CrossRef]
  170. Parker, E.M.; Hutchison, M.; Blumer, J.L. The pharmacokinetics of meropenem in infants and children: A population analysis. J. Antimicrob. Chemother. 1995, 36 (Suppl. A), 63–71. [Google Scholar] [CrossRef]
  171. Pettit, R.S.; Neu, N.; Cies, J.J.; Lapin, C.; Muhlebach, M.S.; Novak, K.J.; Nguyen, S.T.; Saiman, L.; Nicolau, D.P.; Kuti, J.L. Population pharmacokinetics of meropenem administered as a prolonged infusion in children with cystic fibrosis. J. Antimicrob. Chemother. 2016, 71, 189–195. [Google Scholar] [CrossRef]
  172. Du, X.; Li, C.; Kuti, J.L.; Nightingale, C.H.; Nicolau, D.P. Population pharmacokinetics and pharmacodynamics of meropenem in pediatric patients. J. Clin. Pharmacol. 2006, 46, 69–75. [Google Scholar] [CrossRef]
  173. Ikawa, K.; Morikawa, N.; Ikeda, K.; Miki, M.; Kobayashi, M. Population pharmacokinetics and pharmacodynamics of meropenem in Japanese pediatric patients. J. Infect. Chemother. 2010, 16, 139–143. [Google Scholar] [CrossRef]
  174. Monogue, M.L.; Pettit, R.S.; Muhlebach, M.; Cies, J.J.; Nicolau, D.P.; Kuti, J.L. Population Pharmacokinetics and Safety of Ceftolozane-Tazobactam in Adult Cystic Fibrosis Patients Admitted with Acute Pulmonary Exacerbation. Antimicrob. Agents Chemother. 2016, 60, 6578–6584. [Google Scholar] [CrossRef]
  175. Zhanel, G.G.; Chung, P.; Adam, H.; Zelenitsky, S.; Denisuik, A.; Schweizer, F.; Lagacé-Wiens, P.R.S.; Rubinstein, E.; Gin, A.S.; Walkty, A.; et al. Ceftolozane/tazobactam: A novel cephalosporin/β-lactamase inhibitor combination with activity against multidrug-resistant gram-negative bacilli. Drugs 2014, 74, 31–51. [Google Scholar] [CrossRef]
  176. Chandorkar, G.; Xiao, A.; Mouksassi, M.-S.; Hershberger, E.; Krishna, G. Population pharmacokinetics of ceftolozane/tazobactam in healthy volunteers, subjects with varying degrees of renal function and patients with bacterial infections. J. Clin. Pharmacol. 2015, 55, 230–239. [Google Scholar] [CrossRef] [PubMed]
  177. Singh, S.B.; McLearn-Montz, A.J.; Milavetz, F.; Gates, L.K.; Fox, C.; Murry, L.T.; Sabus, A.; Porterfield, H.S.; Fischer, A.J. Pathogen acquisition in patients with cystic fibrosis receiving ivacaftor or lumacaftor/ivacaftor. Pediatr. Pulmonol. 2019, 54, 1200–1208. [Google Scholar] [CrossRef] [PubMed]
  178. Ledger, E.L.; Smith, D.J.; Teh, J.J.; Wood, M.E.; Whibley, P.E.; Morrison, M.; Goldberg, J.B.; Reid, D.W.; Wells, T.J. Impact of CFTR Modulation on Pseudomonas aeruginosa Infection in People with Cystic Fibrosis. J. Infect. Dis. 2024, 230, e536–e547. [Google Scholar] [CrossRef]
  179. Guimbellot, J.S.; Baines, A.; Paynter, A.; Heltshe, S.L.; VanDalfsen, J.; Jain, M.; Rowe, S.M.; Sagel, S.D.; GOAL-e2 Investigators. Long term clinical effectiveness of ivacaftor in people with the G551D CFTR mutation. J. Cyst. Fibros. 2021, 20, 213–219. [Google Scholar] [CrossRef]
  180. Deeks, E.D. Ivacaftor: A review of its use in patients with cystic fibrosis. Drugs 2013, 73, 1595–1604. [Google Scholar] [CrossRef]
  181. Deeks, E.D. Lumacaftor/Ivacaftor: A Review in Cystic Fibrosis. Drugs 2016, 76, 1191–1201. [Google Scholar] [CrossRef]
  182. Markussen, T.; Marvig, R.L.; Gómez-Lozano, M.; Aanæs, K.; Burleigh, A.E.; Høiby, N.; Johansen, H.K.; Molin, S.; Jelsbak, L. Environmental heterogeneity drives within-host diversification and evolution of Pseudomonas aeruginosa. mBio 2014, 5, e01592-14. [Google Scholar] [CrossRef]
  183. Pereira, S.G.; Cardoso, O. Mobile genetic elements of Pseudomonas aeruginosa isolates from hydrotherapy facility and respiratory infections. Clin. Microbiol. Infect. 2014, 20, O203–O206. [Google Scholar] [CrossRef]
  184. Waine, D.J.; Honeybourne, D.; Smith, E.G.; Whitehouse, J.L.; Dowson, C.G. Cross-sectional and longitudinal multilocus sequence typing of Pseudomonas aeruginosa in cystic fibrosis sputum samples. J. Clin. Microbiol. 2009, 47, 3444–3448. [Google Scholar] [CrossRef]
  185. Mowat, E.; Paterson, S.; Fothergill, J.L.; Wright, E.A.; Ledson, M.J.; Walshaw, M.J.; Brockhurst, M.A.; Winstanley, C. Pseudomonas aeruginosa population diversity and turnover in cystic fibrosis chronic infections. Am. J. Respir. Crit. Care Med. 2011, 183, 1674–1679. [Google Scholar] [CrossRef]
  186. Schick, A.; Shewaramani, S.; Kassen, R. Genomics of Diversification of Pseudomonas aeruginosa in Cystic Fibrosis Lung-like Conditions. Genome Biol. Evol. 2022, 14, evac074. [Google Scholar] [CrossRef] [PubMed]
  187. Martin, L.W.; Gray, A.R.; Brockway, B.; Lamont, I.L. Pseudomonas aeruginosa is oxygen-deprived during infection in cystic fibrosis lungs, reducing the effectiveness of antibiotics. FEMS Microbiol. Lett. 2023, 370, fnad076. [Google Scholar] [CrossRef] [PubMed]
  188. Faure, E.; Kwong, K.; Nguyen, D. Pseudomonas aeruginosa in Chronic Lung Infections: How to Adapt Within the Host? Front. Immunol. 2018, 9, 2416. [Google Scholar] [CrossRef]
  189. Gibson, R.L.; Burns, J.L.; Ramsey, B.W. Pathophysiology and management of pulmonary infections in cystic fibrosis. Am. J. Respir. Crit. Care Med. 2003, 168, 918–951. [Google Scholar] [CrossRef]
  190. Sunman, B.; Emiralioglu, N.; Hazirolan, G.; Şener, B.; Ozsezen, B.; Tural, D.A.; Buyuksahin, H.N.; Guzelkas, I.; Yalcin, E.; Dogru, D.; et al. Effectiveness of different eradication treatment protocols for new-onset Pseudomonas aeruginosa in children with cystic fibrosis. Pediatr. Pulmonol. 2022, 57, 1456–1465. [Google Scholar] [CrossRef]
  191. Akkerman-Nijland, A.M.; Akkerman, O.W.; Grasmeijer, F.; Hagedoorn, P.; Frijlink, H.W.; Rottier, B.L.; Koppelman, G.H.; Touw, D.J. The pharmacokinetics of antibiotics in cystic fibrosis. Expert. Opin. Drug Metab. Toxicol. 2021, 17, 53–68. [Google Scholar] [CrossRef]
  192. Cogen, J.D.; Sanders, D.B.; Slaven, J.E.; Faino, A.V.; Somayaji, R.; Gibson, R.L.; Hoffman, L.R.; Ren, C.L. Antibiotic Regimen Changes during Cystic Fibrosis Pediatric Pulmonary Exacerbation Treatment. Ann. Am. Thorac. Soc. 2023, 20, 1293–1298. [Google Scholar] [CrossRef]
  193. Lewis, R.H.; Sharpe, J.P.; Swanson, J.M.; Fabian, T.C.; Croce, M.A.; Magnotti, L.J. Reinventing the wheel: Impact of prolonged antibiotic exposure on multidrug-resistant ventilator-associated pneumonia in trauma patients. J. Trauma. Acute Care Surg. 2018, 85, 256–262. [Google Scholar] [CrossRef]
  194. Zhao, A.; Sun, J.; Liu, Y. Understanding bacterial biofilms: From definition to treatment strategies. Front. Cell. Inf. Microbio. 2023, 13, 1137947-23. [Google Scholar] [CrossRef]
  195. Ciofu, O.; Rojo-Molinero, E.; Macià, M.D.; Oliver, A. Antibiotic treatment of biofilm infections. APMIS 2017, 125, 304–319. [Google Scholar] [CrossRef]
  196. Macia, M.D.; Rojo-Molinero, E.; Oliver, A. Antimicrobial susceptibility testing in biofilm-growing bacteria. Clin. Microbiol. Infect. 2014, 20, 981–990. [Google Scholar] [CrossRef] [PubMed]
  197. Kwong, K.; Benedetti, A.; Yau, Y.; Waters, V.; Nguyen, D. Failed Eradication Therapy of New-Onset Pseudomonas aeruginosa Infections in Children with Cystic Fibrosis Is Associated with Bacterial Resistance to Neutrophil Functions. J. Infect. Dis. 2022, 225, 1886–1895. [Google Scholar] [CrossRef] [PubMed]
  198. Garcia-Clemente, M.; de la Rosa, D.; Máiz, L.; Girón, R.; Blanco, M.; Olveira, C.; Canton, R.; Martinez-García, M.A. Impact of Pseudomonas aeruginosa Infection on Patients with Chronic Inflammatory Airway Diseases. J. Clin. Med. 2020, 9, 3800. [Google Scholar] [CrossRef] [PubMed]
  199. Emerson, J.; Rosenfeld, M.; McNamara, S.; Ramsey, B.; Gibson, R.L. Pseudomonas aeruginosa and other predictors of mortality and morbidity in young children with cystic fibrosis. Pediatr. Pulmonol. 2002, 34, 91–100. [Google Scholar] [CrossRef]
  200. Stanford, G.E.; Dave, K.; Simmonds, N.J. Pulmonary Exacerbations in Adults with Cystic Fibrosis: A Grown-up Issue in a Changing Cystic Fibrosis Landscape. CHEST 2021, 159, 93–102. [Google Scholar] [CrossRef]
  201. Goss, C.H. Acute Pulmonary Exacerbations in Cystic Fibrosis. Semin. Respir. Crit. Care Med. 2019, 40, 792–803. [Google Scholar] [CrossRef]
  202. Liou, T.G.; Adler, F.R.; Fitzsimmons, S.C.; Cahill, B.C.; Hibbs, J.R.; Marshall, B.C. Predictive 5-year survivorship model of cystic fibrosis. Am. J. Epidemiol. 2001, 153, 345–352. [Google Scholar] [CrossRef]
  203. Mayer-Hamblett, N.; Rosenfeld, M.; Emerson, J.; Goss, C.H.; Aitken, M.L. Developing cystic fibrosis lung transplant referral criteria using predictors of 2-year mortality. Am. J. Respir. Crit. Care Med. 2002, 166, 1550–1555. [Google Scholar] [CrossRef]
  204. Bhatt, J.M. Treatment of pulmonary exacerbations in cystic fibrosis. Eur. Respir. Rev. 2013, 22, 205–216. [Google Scholar] [CrossRef]
  205. Hahn, A.; Sami, I.; Chaney, H.; Koumbourlis, A.C.; Del Valle Mojica, C.; Cochrane, C.; Chan, B.K.; Koff, J.L. Bacteriophage Therapy for Pan-Drug-Resistant Pseudomonas aeruginosa in Two Persons with Cystic Fibrosis. J. Investig. Med. High. Impact Case Rep. 2023, 11. [Google Scholar] [CrossRef]
  206. Yang, Q.; Le, S.; Zhu, T.; Wu, N. Regulations of phage therapy across the world. Front. Microbiol. 2023, 14, 1250848. [Google Scholar] [CrossRef] [PubMed]
  207. Żaczek, M.; Weber-Dąbrowska, B.; Międzybrodzki, R.; Łusiak-Szelachowska, M.; Górski, A. Phage Therapy in Poland—A Centennial Journey to the First Ethically Approved Treatment Facility in Europe. Front. Microbiol. 2020, 11, 1056. [Google Scholar] [CrossRef] [PubMed]
  208. Tao, S.; Chen, H.; Li, N.; Liang, W. The Application of the CRISPR-Cas System in Antibiotic Resistance. Infect. Drug Resist. 2022, 15, 4155–4168. [Google Scholar] [CrossRef]
  209. Maule, G.; Arosio, D.; Cereseto, A. Gene Therapy for Cystic Fibrosis: Progress and Challenges of Genome Editing. Int. J. Mol. Sci. 2020, 21, 3903. [Google Scholar] [CrossRef]
  210. Wang, G. Genome Editing for Cystic Fibrosis. Cells 2023, 12, 1555. [Google Scholar] [CrossRef]
  211. Mayorga-Ramos, A.; Zúñiga-Miranda, J.; Carrera-Pacheco, S.E.; Barba-Ostria, C.; Guamán, L.P. CRISPR-Cas-Based Antimicrobials: Design, Challenges, and Bacterial Mechanisms of Resistance. ACS Infect. Dis. 2023, 9, 1283–1302. [Google Scholar] [CrossRef]
  212. Martin, I.; Waters, V.; Grasemann, H. Approaches to Targeting Bacterial Biofilms in Cystic Fibrosis Airways. Int. J. Mol. Sci. 2021, 22, 2155. [Google Scholar] [CrossRef]
  213. Roy, R.; Tiwari, M.; Donelli, G.; Tiwari, V. Strategies for combating bacterial biofilms: A focus on anti-biofilm agents and their mechanisms of action. Virulence 2018, 9, 522–554. [Google Scholar] [CrossRef]
  214. Louis, M.; Clamens, T.; Tahrioui, A.; Desriac, F.; Rodrigues, S.; Rosay, T.; Harmer, N.; Diaz, S.; Barreau, M.; Racine, P.-J.; et al. Pseudomonas aeruginosa Biofilm Dispersion by the Human Atrial Natriuretic Peptide. Adv. Sci. 2022, 9, e2103262. [Google Scholar] [CrossRef]
Table 1. Clinical Trial Phases.
Table 1. Clinical Trial Phases.
Pre-ClinicalPhase IPhase IIPhase IIIPhase IV
Involves laboratory and animal studies to evaluate the safety, biological effects, and therapeutic potential of a new drug or treatment.
Aims
Identifies possible toxicities and pharmacokinetic profiles.
Determines if a drug is suitable for human trials.
The initial phase of human testing.
Typically involves 10–80 healthy volunteers.
Aims
Assess the safety of a new drug.
Establish safe dosage levels.
Observe any early signs of adverse effects.
Includes a larger group of patients (100–300) with the condition the drug aims to treat.
Aims
Evaluates the drug’s effectiveness.
Refines the dosing regimen.
Continues to assess its safety profile.
Conducted with larger patient populations (1000–3000+).
Aims
Verifies the treatment’s efficacy.
Monitor for side effects.
Compares the new drug to existing standard therapies.
Provides comprehensive data for regulatory approval.
Started after the drug approval and is available to the public.
Aims
Focuses on long-term safety, effectiveness.
Identifies any rare side effects that may emerge with widespread use of a drug.
Ensures the drug’s continued safety in a diverse patient population.
Table 2. Phase IV trials for the treatment of Pa infections in pwCF.
Table 2. Phase IV trials for the treatment of Pa infections in pwCF.
NoStudy TitleAccession NumberTreatmentsNStart DateCompletion DatePrimary Measures (Endpoint)Main FindingsTime Frame
1Long Term Safety of Tobramycin Inhalation Powder in Patients with Cystic FibrosisNCT01519661
PMID(27435882)
Tobramycin inhalation powder (TIP)15701-201201-2014The proportion of participants experiencing treatment-emergent adverse events (AEs), serious adverse events (SAEs), and deaths.No new safety concerns emerged, and the intervention maintained its effectiveness throughout the study period337 days
2Ext. Long-term Safety Study in CF Patients: Single Arm TIP 1NCT01775137Tobramycin inhalation powder (TIP)4502-201311-2014To determine the number of participants with AEs, SAEs, and AEs/SAEs leading to discontinuation of the study drug, and deaths across 12 treatment cycles.The adverse event profile was similar to the core study.673 days
3IV Colistin for Pulmonary Exacerbations: Improving Safety and
Efficacy
NCT02918409Colistin
Tobramycin
5108-201611-2021To evaluate the efficacy and safety of standard colistin dosing (2.5 mg/kg/day, administered three times daily, or TID) compared with a pharmacokinetically (PK)-adjusted colistin regimen administered twice daily (BID) at 5 mg/kg/day in adult cystic fibrosis (CF) patients with acute pulmonary exacerbation (APE).
To assess the efficacy and safety of PK-adjusted BID colistin dosing compared with standard once-daily tobramycin dosing (8–10 mg/kg) in adult CF patients with APE.
Notable safety concerns were observed. IV tobramycin resulted in more renal toxicity events compared with other study arms.
Both tobramycin and colistin resulted in an improvement in lung function.
Up to 14 days
4Ease of Use and Microbial Contamination of Tobramycin
Inhalation Powder (TIP) Versus Nebulised Tobramycin
Inhalation Solution (TIS) and Nebulised Colistimethate (COLI)
NCT01844778
PMID(28614995)
Tobramycin inhalation powder (TIP)
Tobramycin inhalation solution (TIS)
Colistimethate
6008-201310-2015To evaluate the mean administration time of TIP using the T-326 inhaler compared with the total administration time of COLI or TIS.The T-326 inhaler for TIP administration demonstrated ease of use, reduced total delivery time, and significantly lowered contamination rates compared with nebulizers.Days 1, 28, 57, 84, 112
5Population Pharmacokinetics of Prolonged Infusion Meropenem in Cystic Fibrosis (CF) ChildrenNCT01429259
PMID(26416780)
Meropenem3002-201201-2014To determine total body clearance of meropenem at different concentrations by analyzing the pharmacokinetics of the study population.
To determine the central compartment volume of meropenem at different concentrations by analyzing the pharmacokinetics of the study population.
Cystic fibrosis (CF) children exhibited higher meropenem clearance compared with previously reported values in non-CF children.
Prolonged infusion improved drug exposure against pathogens with minimum inhibitory concentrations (MICs) ≥ 1 mg/L.
8-h dosing interval after 3rd meropenem dose /14–21 days
6Population Pharmacokinetics and Safety of Intravenous
Ceftolozane/Tazobactam in Adult Cystic Fibrosis Patients
NCT02421120
PMID(27550351)
Ceftolozane/Tazobactam2109-201510-2016To evaluate the clearance of ceftolozane and tazobactam during an 8 h dosing interval.
To assess the volume of distribution of ceftolozane and tazobactam over the same 8 h dosing interval.
Ceftolozane-tazobactam demonstrated good tolerability in this study.
Ceftolozane and tazobactam clearance in cystic fibrosis (CF) patients was comparable to non-CF adult populations.
AEs associated with ceftolozane-tazobactam were mild and potentially attributable to co-administered medications in most cases.
0, 1–1.08, 1.25–1.5, 2–3, 4–5, and 7–8 h after start of final dose/3 days
7Testing the Effect of Adding Chronic Oral Azithromycin to Inhaled Tobramycin in People with Cystic Fibrosis (CF)NCT02677701Azithromycin plus inhaled tobramycin11910-201602-2020To assess the effects of long-term oral azithromycin combined with inhaled tobramycin in adolescents and adults with CF and chronic Pa infectionsNo significant improvements in lung function were detected with the addition of Azithromycin.
6 weeks
8Scandinavian Cystic Fibrosis Azithromycin StudyNCT00411736Azithromycin plus inhaled colistin and ciprofloxacin4505-200803-2014To assess if adding low-dose azithromycin to standard inhaled colistin and oral ciprofloxacin for treating intermittent Pseudomonas airway infections can delay recurrence and prevent progression to chronic airway infection.No results reported.Up to 5 Years
9Comparison of 2 Treatment Regimens for Eradication of P Aeruginosa Infection in Children with Cystic FibrosisNCT01400750Tobramycin inhalation solution (TIS)
Inhaled colistimethate sodium plus oral ciprofloxacin (CC)
6108-200105-2011To compare the ability of two regimens to successfully achieve Pa eradication at the end of their treatment plans.No results reported.1 months for TIS
3 months for CC
10Aztreonam for Inhalation Solution (AZLI) for the Treatment of Exacerbations of Cystic FibrosisNCT02894684Aztreonam inhalation solution (AZLI)1601-201709-2019To assess the clinical efficacy of AZLI in treating acute pulmonary exacerbations.No results reported.14 days
The data were retrieved on 27 September 2024 from ClinicalTrials.gov using the search terms “Cystic fibrosis” and “Pseudomonas infection”. Only completed phase IV trials were included. 1 An extension of study 1 at this table.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alqasmi, M. Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials. J. Clin. Med. 2024, 13, 6530. https://doi.org/10.3390/jcm13216530

AMA Style

Alqasmi M. Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials. Journal of Clinical Medicine. 2024; 13(21):6530. https://doi.org/10.3390/jcm13216530

Chicago/Turabian Style

Alqasmi, Mohammed. 2024. "Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials" Journal of Clinical Medicine 13, no. 21: 6530. https://doi.org/10.3390/jcm13216530

APA Style

Alqasmi, M. (2024). Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials. Journal of Clinical Medicine, 13(21), 6530. https://doi.org/10.3390/jcm13216530

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop