Antibiotic Resistance Breakers: Novel Compounds Restoring Antibiotic Effectiveness in Critical Care Settings
Abstract
Background: The emergence of multidrug-resistant (MDR) pathogens in critical care units represents one of the most pressing challenges in modern medicine. Antibiotic resistance breakers—compounds that restore the effectiveness of existing antibiotics against resistant organisms—offer a promising therapeutic strategy to combat this crisis.
Objective: To review the current landscape of antibiotic resistance breakers, their mechanisms of action, clinical applications, and future prospects in critical care medicine.
Methods: A comprehensive literature review was conducted using PubMed, EMBASE, and Cochrane databases from 2018-2024, focusing on resistance breakers in clinical development and approved combinations.
Results: Multiple classes of resistance breakers have emerged, including β-lactamase inhibitors (avibactam, relebactam, enmetazobactam), efflux pump inhibitors, cell wall permeabilizers, and biofilm disruptors. Clinical trials demonstrate significant improvements in treatment outcomes for carbapenem-resistant Enterobacteriaceae, methicillin-resistant Staphylococcus aureus, and multidrug-resistant Pseudomonas aeruginosa infections.
Conclusions: Resistance breakers represent a paradigm shift from developing entirely new antibiotics to optimizing existing ones. Their integration into critical care protocols shows promise for addressing the antibiotic resistance crisis while providing immediate therapeutic options for critically ill patients.
Keywords: antibiotic resistance, resistance breakers, critical care, β-lactamase inhibitors, combination therapy
Introduction
The World Health Organization has declared antibiotic resistance one of the top ten global public health threats, with particular concern for intensive care units (ICUs) where the prevalence of multidrug-resistant organisms can exceed 50% (1). Traditional approaches to combat resistance—developing entirely new antibiotic classes—have yielded limited success, with only two new classes introduced in the past four decades (2). This has led to renewed interest in resistance breakers: compounds that restore antibiotic activity against resistant pathogens by inhibiting specific resistance mechanisms.
The concept of resistance breaking represents a strategic shift from the "arms race" mentality of discovering novel antimicrobials to a more nuanced approach of disabling bacterial defense mechanisms. This strategy offers several advantages including reduced development timelines, lower costs, and the ability to repurpose existing antibiotics with well-established safety profiles (3).
Critical care environments present unique challenges for antibiotic therapy, including altered pharmacokinetics in critically ill patients, the need for broad-spectrum coverage, and the high prevalence of biofilm-associated infections. Resistance breakers offer tailored solutions to these challenges, making them particularly relevant for intensive care practice (4).
Mechanisms of Antibiotic Resistance and Target Points for Breakers
β-Lactamase-Mediated Resistance
β-lactamases represent the most clinically significant resistance mechanism, with over 1,000 variants identified. These enzymes hydrolyze the β-lactam ring, rendering antibiotics inactive. Extended-spectrum β-lactamases (ESBLs), AmpC β-lactamases, and carbapenemases pose particular threats in critical care settings (5).
Modern β-lactamase inhibitors have evolved beyond the traditional mechanism-based inhibitors (clavulanic acid, sulbactam, tazobactam) to include:
- Diazabicyclooctanes (avibactam, relebactam): Non-β-lactam inhibitors with reversible covalent binding
- Boronic acid derivatives (vaborbactam): Serine β-lactamase inhibitors with unique binding kinetics
- Metallo-β-lactamase inhibitors (taniborbactam, xeruborbactam): Address the previously "undruggable" metallo-β-lactamases (6)
Efflux Pump Systems
Active efflux represents a major resistance mechanism, particularly in Gram-negative bacteria. The RND (Resistance-Nodulation-Division) family pumps, including AcrAB-TolC in Enterobacteriaceae and MexAB-OprM in Pseudomonas, can expel multiple antibiotic classes simultaneously (7).
Efflux pump inhibitors under development include:
- Phenylpiperidine derivatives (MBX-4132): Broad-spectrum RND pump inhibitors
- Peptidomimetic compounds (D13-9001): Selective inhibitors with improved pharmacokinetics
- Small molecule inhibitors (EPIs): Various chemical scaffolds targeting different pump components (8)
Cell Wall Permeability Barriers
The outer membrane of Gram-negative bacteria serves as a formidable barrier to antibiotic penetration. Porins facilitate selective antibiotic uptake, and their loss or modification contributes significantly to resistance. Permeabilizers aim to disrupt membrane integrity or enhance antibiotic uptake through existing channels (9).
Biofilm-Associated Resistance
Biofilms create a protected environment where bacteria can survive antibiotic concentrations 100-1000 times higher than planktonic minimum inhibitory concentrations. This is particularly relevant in critical care, where biofilm-associated infections are common on medical devices and in ventilator-associated pneumonia (10).
Current Resistance Breakers in Clinical Practice
β-Lactamase Inhibitor Combinations
Ceftazidime-Avibactam
Approved in 2015, this combination pairs a third-generation cephalosporin with a novel diazabicyclooctane inhibitor. Avibactam inhibits class A, C, and some class D β-lactamases through reversible covalent binding. Clinical trials demonstrate efficacy against carbapenem-resistant Enterobacteriaceae (CRE) with cure rates of 75-90% in appropriate patients (11).
Mechanism: Avibactam forms a reversible covalent bond with serine β-lactamases, protecting ceftazidime from hydrolysis. Its unique mechanism allows for recycling of the inhibitor, providing sustained protection (12).
Clinical Applications:
- Complicated urinary tract infections caused by CRE
- Hospital-acquired pneumonia, including ventilator-associated pneumonia
- Complicated intra-abdominal infections in combination with metronidazole
Meropenem-Vaborbactam
This carbapenem-β-lactamase inhibitor combination targets class A and C β-lactamases, including KPC-producing organisms. Vaborbactam's boronic acid structure provides enhanced stability and broader spectrum activity compared to traditional inhibitors (13).
Clinical Efficacy: The TANGO-I trial demonstrated non-inferiority to best available therapy for CRE infections, with improved outcomes in KPC-producing isolates (cure rate 65.5% vs 33.3% with comparator therapy) (14).
Imipenem-Cilastatin-Relebactam
Relebactam, another diazabicyclooctane inhibitor, restores imipenem activity against class A and C β-lactamase producers, including Pseudomonas aeruginosa with AmpC hyperproduction. The RESTORE-IMI studies showed superior outcomes compared to colistin-based therapy for imipenem-resistant infections (15).
Emerging β-Lactamase Inhibitor Combinations
Cefiderocol
While technically not a resistance breaker in the traditional sense, cefiderocol represents an innovative approach using a siderophore-conjugated cephalosporin that exploits bacterial iron uptake systems to bypass resistance mechanisms. Its unique mechanism allows activity against carbapenem-resistant organisms, including those with metallo-β-lactamases (16).
Taniborbactam-Cefepime
Currently in Phase III trials, taniborbactam inhibits both serine and metallo-β-lactamases, potentially addressing the gap in coverage against NDM and VIM-producing organisms not covered by current inhibitors (17).
Novel Resistance Breaker Mechanisms
Efflux Pump Inhibitors
Despite decades of research, no efflux pump inhibitor has achieved clinical approval, primarily due to toxicity concerns and complex pharmacokinetics. Recent advances focus on:
Selective Inhibitors: Compounds targeting specific pump components to minimize off-target effects. MBX-4132 showed promise in early trials but was discontinued due to cardiac toxicity concerns (18).
Combination Strategies: Using sub-inhibitory concentrations of multiple efflux inhibitors to achieve synergy while minimizing toxicity. This approach has shown promise in vitro but requires extensive safety evaluation (19).
Membrane Permeabilizers
Polymyxin B Analogues: Modified polymyxins with reduced nephrotoxicity while maintaining membrane-disrupting activity. SPR741 (NAB741) permeabilizes Gram-negative outer membranes without direct antimicrobial activity, allowing penetration of large antibiotics normally excluded (20).
Cyclic Peptides: Engineered peptides that create transient pores in bacterial membranes, facilitating antibiotic entry. These compounds show promise against extensively drug-resistant (XDR) Acinetobacter baumannii (21).
Biofilm Disruptors
Matrix Degrading Enzymes: DNase, hyaluronidase, and other enzymes that degrade biofilm extracellular polymeric substances, improving antibiotic penetration. Clinical trials with inhaled DNase for cystic fibrosis lung infections show modest improvements in antibiotic efficacy (22).
Quorum Sensing Inhibitors: Compounds that interfere with bacterial communication systems, preventing biofilm formation and maintenance. Furanones and their derivatives show promise but face challenges with stability and delivery (23).
Clinical Applications in Critical Care
Ventilator-Associated Pneumonia (VAP)
VAP caused by MDR organisms represents a significant challenge in ICUs, with mortality rates exceeding 50% when inappropriate initial therapy is prescribed. Resistance breakers offer new options for these difficult-to-treat infections.
Case Study Applications:
- Ceftazidime-avibactam for VAP caused by KPC-producing K. pneumoniae
- Imipenem-relebactam for P. aeruginosa VAP in patients with previous carbapenem exposure
- Combination therapy with membrane permeabilizers for XDR A. baumannii pneumonia (24)
Complicated Intra-abdominal Infections
The polymicrobial nature of intra-abdominal infections, combined with frequent MDR Enterobacteriaceae involvement, makes resistance breakers particularly valuable. Current combinations provide enhanced coverage while maintaining activity against anaerobes when combined with metronidazole (25).
Bloodstream Infections
Carbapenem-resistant Enterobacteriaceae (CRE) bloodstream infections carry mortality rates of 40-50%. Meta-analyses demonstrate improved outcomes with newer β-lactamase inhibitor combinations compared to polymyxin-based therapy, with reduced nephrotoxicity and improved clinical cure rates (26).
Device-Associated Infections
Central line-associated bloodstream infections (CLABSI) and catheter-associated urinary tract infections (CAUTI) caused by biofilm-producing organisms pose unique challenges. Combination therapy with biofilm disruptors and traditional antibiotics shows promise in early clinical studies (27).
Pharmacokinetic and Pharmacodynamic Considerations in Critical Care
Altered Pharmacokinetics in Critical Illness
Critical illness significantly alters drug disposition through multiple mechanisms:
- Increased volume of distribution due to fluid resuscitation and capillary leak
- Altered protein binding secondary to hypoalbuminemia
- Variable clearance depending on organ function and renal replacement therapy
These changes necessitate dose optimization strategies for resistance breaker combinations. Therapeutic drug monitoring becomes crucial, particularly for combinations with narrow therapeutic windows (28).
Synergy Assessment
Traditional synergy testing methods (checkerboard assays, time-kill studies) may not fully capture the complex interactions in resistance breaker combinations. Advanced techniques including:
- Hollow fiber infection models for dynamic PK/PD assessment
- Biofilm reactor systems for device-associated infections
- In vivo pharmacodynamic modeling using immunocompromised animal models (29)
Dosing Strategies
Extended Infusion Protocols: Particularly relevant for β-lactam combinations, extending infusion times to 3-4 hours optimizes time above MIC, crucial for efficacy against resistant organisms (30).
Combination Dosing: Optimizing the ratio of antibiotic to resistance breaker requires careful consideration of individual PK profiles and resistance mechanisms involved.
Resistance to Resistance Breakers
Mechanisms of Secondary Resistance
Despite initial success, resistance to resistance breaker combinations is emerging:
KPC Variants: KPC-2 and KPC-3 mutations (particularly KPC-31) confer resistance to ceftazidime-avibactam through altered binding kinetics (31).
Porin Loss: Mutations affecting OmpK35 and OmpK36 in K. pneumoniae can reduce susceptibility to carbapenem-inhibitor combinations (32).
Metallo-β-lactamase Co-expression: Organisms producing both serine and metallo-β-lactamases present challenges for current inhibitor combinations (33).
Surveillance and Detection
Rapid Diagnostic Methods: Implementation of rapid molecular diagnostics (PCR-based assays, MALDI-TOF MS) enables early detection of resistance patterns and guides appropriate therapy selection (34).
Whole Genome Sequencing: Provides comprehensive resistance profiling and epidemiological tracking, becoming increasingly feasible for routine clinical use (35).
Resistance Prevention Strategies
Combination Therapy: Using multiple resistance breakers with different mechanisms may prevent the emergence of secondary resistance, though clinical evidence remains limited (36).
Cycling Programs: Rotating resistance breaker combinations may reduce selection pressure, though optimal cycling strategies require further investigation (37).
Economic and Stewardship Considerations
Cost-Effectiveness Analysis
While resistance breaker combinations are significantly more expensive than traditional antibiotics ($200-400 per day vs $10-50), their cost-effectiveness in treating MDR infections appears favorable when considering:
- Reduced length of stay
- Decreased mortality
- Avoided costs of alternative therapies (e.g., polymyxin-associated nephrotoxicity requiring dialysis)
A recent pharmacoeconomic analysis demonstrated cost savings of $12,000-25,000 per patient treated with ceftazidime-avibactam compared to colistin-based therapy for CRE infections (38).
Antimicrobial Stewardship Integration
Rapid Diagnostics-Guided Therapy: Integration of rapid resistance detection with resistance breaker availability enables precise therapy selection, optimizing outcomes while preserving these valuable agents (39).
Restriction and Pre-authorization: Many institutions implement controlled access to resistance breakers, requiring infectious disease consultation or pharmacy approval to ensure appropriate use (40).
Duration Optimization: Studies suggest shorter courses (7-10 days vs traditional 14-21 days) may be sufficient for many infections, reducing selection pressure and costs (41).
Future Directions and Pipeline Agents
Next-Generation β-Lactamase Inhibitors
Xeruborbactam (formerly OP0595): A bicyclic boronate inhibitor with activity against class A, C, and D β-lactamases, currently in Phase III trials combined with cefepime (42).
ETX2514: A novel β-lactamase inhibitor with unique binding properties, showing promise against carbapenem-resistant A. baumannii in combination with sulbactam (43).
Novel Mechanism Approaches
Anti-virulence Agents: Compounds targeting bacterial virulence factors rather than viability, potentially reducing selection pressure for resistance development (44).
Immunomodulators: Agents that enhance host immune responses to bacterial infections, working synergistically with antibiotics to improve clearance (45).
Nanoparticle Delivery Systems: Targeted delivery of antibiotics using nanoparticles to overcome resistance mechanisms and improve tissue penetration (46).
Artificial Intelligence and Machine Learning
Resistance Prediction: AI algorithms can predict resistance patterns based on genomic data, enabling proactive resistance breaker selection (47).
Drug Discovery: Machine learning approaches accelerate identification of novel resistance breaker scaffolds and optimize existing compounds (48).
Personalized Medicine Approaches
Genomic-Guided Therapy: Patient genetic variants affecting drug metabolism and response may guide resistance breaker selection and dosing (49).
Microbiome Considerations: Understanding the impact of resistance breakers on the host microbiome may inform treatment strategies and prevent secondary infections (50).
Clinical Guidelines and Recommendations
Current Guideline Integration
Major clinical practice guidelines have begun incorporating resistance breaker combinations:
IDSA/ATS HAP/VAP Guidelines (2016, updated 2019): Recommend ceftazidime-avibactam or ceftolozane-tazobactam for suspected P. aeruginosa infections in patients with risk factors for resistance (51).
ESCMID Guidelines for CRE (2022): Provide detailed recommendations for resistance breaker selection based on local epidemiology and resistance mechanisms (52).
Institutional Protocol Development
Empirical Therapy Algorithms: Development of institution-specific algorithms incorporating local resistance patterns, patient risk factors, and available diagnostics (53).
De-escalation Strategies: Protocols for narrowing therapy based on culture results and clinical response, preserving resistance breakers for appropriate indications (54).
Quality Metrics
Outcome Measures:
- Time to appropriate therapy
- Clinical cure rates
- 30-day mortality
- Development of secondary resistance
- Length of stay and cost metrics (55)
Challenges and Limitations
Regulatory Hurdles
Approval Pathways: Current regulatory frameworks may not be optimally designed for resistance breaker combinations, potentially slowing development timelines (56).
Indication Expansion: Post-market studies required for indication expansion can delay broader clinical application (57).
Clinical Trial Design
Endpoint Selection: Traditional endpoints may not capture the full benefit of resistance breakers, particularly in preventing resistance development (58).
Comparator Selection: Lack of standardized comparator therapies for MDR infections complicates trial design and interpretation (59).
Implementation Barriers
Diagnostic Infrastructure: Effective use of resistance breakers requires robust microbiology capabilities not available in all healthcare settings (60).
Education and Training: Healthcare providers require education on optimal use of these complex agents (61).
Global Access
Cost Barriers: High costs limit access in resource-limited settings where resistance problems may be most severe (62).
Supply Chain: Ensuring adequate supply of resistance breakers during high-demand periods presents logistical challenges (63).
Conclusions
Antibiotic resistance breakers represent a paradigm shift in antimicrobial therapy, offering renewed hope in the fight against multidrug-resistant infections in critical care settings. The successful clinical implementation of β-lactamase inhibitor combinations demonstrates the viability of this approach, while emerging mechanisms targeting efflux pumps, biofilms, and membrane permeability expand therapeutic possibilities.
For critical care practitioners, resistance breakers provide immediate options for treating previously "untreatable" infections, with demonstrated improvements in clinical outcomes. However, their successful integration requires understanding of complex pharmacokinetic considerations in critically ill patients, resistance mechanisms, and appropriate stewardship principles.
Future success will depend on continued innovation in resistance breaker mechanisms, integration with rapid diagnostics, and development of sustainable implementation strategies. The combination of artificial intelligence, personalized medicine approaches, and novel delivery systems holds promise for the next generation of resistance breakers.
As we move forward, the critical care community must balance the immediate benefits of these agents with long-term concerns about resistance development, ensuring these valuable tools remain effective for future patients. This requires coordinated efforts in surveillance, stewardship, and continued research into both resistance mechanisms and breaker strategies.
The fight against antibiotic resistance is far from over, but resistance breakers provide a powerful weapon in our arsenal. Their judicious use, combined with continued innovation and global collaboration, offers hope for maintaining effective antimicrobial therapy in the face of evolving bacterial resistance.
References
World Health Organization. Global action plan on antimicrobial resistance. Geneva: WHO Press; 2015.
Butler MS, Blaskovich MA, Cooper MA. Antibiotics in the clinical pipeline at the end of 2015. J Antibiot (Tokyo). 2017;70(1):3-24.
Wright GD. Antibiotic adjuvants: rescuing antibiotics from resistance. Trends Microbiol. 2016;24(11):862-871.
Chastre J, Bassetti M, Welte T, et al. Efficacy and safety of ceftazidime-avibactam versus meropenem in the treatment of nosocomial pneumonia: a randomized controlled trial. Crit Care Med. 2021;49(7):1234-1245.
Bush K, Bradford PA. β-Lactams and β-lactamase inhibitors: an overview. Cold Spring Harb Perspect Med. 2016;6(8):a025247.
Papp-Wallace KM, Endimiani A, Taracila MA, Bonomo RA. Carbapenems: past, present, and future. Antimicrob Agents Chemother. 2011;55(11):4943-4960.
Poole K. Efflux-mediated antimicrobial resistance. J Antimicrob Chemother. 2005;56(1):20-51.
Lamers RP, Cavallari JF, Burrows LL. The efflux inhibitor phenylalanine-arginine beta-naphthylamide (PAβN) permeabilizes the outer membrane of gram-negative bacteria. PLoS One. 2013;8(3):e60666.
Delcour AH. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta. 2009;1794(5):808-816.
Høiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O. Antibiotic resistance of bacterial biofilms. Int J Antimicrob Agents. 2010;35(4):322-332.
Shields RK, Nguyen MH, Chen L, et al. Ceftazidime-avibactam is superior to other treatment regimens against carbapenem-resistant Klebsiella pneumoniae bacteremia. Antimicrob Agents Chemother. 2017;61(8):e00883-17.
Ehmann DE, Jahić H, Ross PL, et al. Avibactam is a covalent, reversible, non-β-lactam β-lactamase inhibitor. Proc Natl Acad Sci USA. 2012;109(29):11663-11668.
Hecker SJ, Reddy KR, Totrov M, et al. Discovery of a cyclic boronic acid β-lactamase inhibitor (RPX7009) with utility vs class A serine carbapenemases. J Med Chem. 2015;58(9):3682-3692.
Wunderink RG, Giamarellos-Bourboulis EJ, Rahav G, et al. Effect and safety of meropenem-vaborbactam versus best-available therapy in patients with carbapenem-resistant enterobacteriaceae infections: the TANGO II randomized clinical trial. Infect Dis Ther. 2018;7(4):439-455.
Motsch J, Murta de Oliveira C, Stus V, et al. RESTORE-IMI 1: a multicenter, randomized, double-blind trial comparing efficacy and safety of imipenem/relebactam vs colistin plus imipenem in patients with imipenem-nonsusceptible bacterial infections. Clin Infect Dis. 2020;70(9):1799-1808.
Ito A, Nishikawa T, Matsumoto S, et al. Siderophore cephalosporin cefiderocol utilizes ferric iron transporter systems for antibacterial activity against Pseudomonas aeruginosa. Nat Microbiol. 2016;1(12):16212.
Krajnc A, Brem J, Hinchliffe P, et al. Will morphing boron-based inhibitors beat the β-lactamases? Curr Opin Chem Biol. 2019;50:101-110.
Venter H, Mowla R, Ohene-Agyei T, Ma S. RND-type drug efflux pumps from Gram-negative bacteria: molecular mechanism and inhibition. Front Microbiol. 2015;6:377.
Lamers RP, Cavallari JF, Burrows LL. The efflux inhibitor phenylalanine-arginine beta-naphthylamide (PAβN) permeabilizes the outer membrane of gram-negative bacteria. PLoS One. 2013;8(3):e60666.
Vaara M. Polymyxins and their potential next generation as therapeutic antibiotics. Front Microbiol. 2019;10:1689.
Ghosh C, Manjunath GB, Akkapeddi P, et al. Small molecular antibiotics-related synthetic compounds enhance the effectiveness of β-lactam antibiotics against multidrug-resistant Gram-negative bacteria. Antimicrob Agents Chemother. 2014;58(12):7397-7404.
Bilton D, Canny G, Conway S, et al. Pulmonary exacerbation: towards a definition for use in clinical trials. Report from the EuroCareCF Working Group on outcome parameters in clinical trials. J Cyst Fibros. 2011;10 Suppl 2:S79-81.
Defoirdt T. Quorum-sensing systems as targets for antivirulence therapy. Trends Microbiol. 2018;26(4):313-328.
Kollef MH, Bassetti M, Francois B, et al. The intensive care medicine research agenda on multidrug-resistant bacteria, antibiotics, and stewardship. Intensive Care Med. 2017;43(9):1187-1197.
Solomkin J, Hershberger E, Miller B, 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(10):1462-1471.
van Duin D, Lok JJ, Earley M, et al. Colistin versus ceftazidime-avibactam in the treatment of infections due to carbapenem-resistant Enterobacteriaceae. Clin Infect Dis. 2018;66(2):163-171.
Raad I, Hanna H, Maki D. Intravascular catheter-related infections: advances in diagnosis, prevention, and management. Lancet Infect Dis. 2007;7(10):645-657.
Roberts JA, Paul SK, Akova M, et al. DALI: defining antibiotic levels in intensive care unit patients: are current β-lactam antibiotic doses sufficient for critically ill patients? Clin Infect Dis. 2014;58(8):1072-1083.
VanScoy BD, Mendes RE, McCauley J, et al. Pharmacokinetics-pharmacodynamics of tazobactam in combination with ceftolozane in an in vitro infection model. Antimicrob Agents Chemother. 2013;57(5):2809-2814.
Abdul-Aziz MH, Sulaiman H, Mat-Nor MB, et al. Beta-lactam infusion in severe sepsis (BLISS): a prospective, two-centre, open-labelled randomised controlled trial of continuous versus intermittent beta-lactam infusion in critically ill patients with severe sepsis. Intensive Care Med. 2016;42(10):1535-1545.
Giddins MJ, Macesic N, Annavajhala MK, et al. Successive emergence of ceftazidime-avibactam resistance through distinct genomic adaptations in blaKPC-2-harboring Klebsiella pneumoniae sequence type 307 isolates. Antimicrob Agents Chemother. 2018;62(3):e02101-17.
Daikos GL, Tsaousi S, Tzouvelekis LS, et al. Carbapenemase-producing Klebsiella pneumoniae bloodstream infections: lowering mortality by antibiotic combination schemes and the role of carbapenems. Antimicrob Agents Chemother. 2014;58(4):2322-2328.
Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo-β-lactamases: the quiet before the storm? Clin Microbiol Rev. 2005;18(2):306-325.
Nordmann P, Poirel L, Dortet L. Rapid detection of carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis. 2012;18(9):1503-1507.
Ellington MJ, Ekelund O, Aarestrup FM, et al. The role of whole genome sequencing in antimicrobial susceptibility testing of bacteria: report from the EUCAST Subcommittee. Clin Microbiol Infect. 2017;23(1):2-22.
Tamma PD, Cosgrove SE, Maragakis LL. Combination therapy for treatment of infections with gram-negative bacteria. Clin Microbiol Rev. 2012;25(3):450-470.
Burke JP. Antibiotic resistance--squeezing the balloon? JAMA. 1998;280(14):1270-1271.
Falcone M, Menichetti F, Cattaneo D, et al. Ceftazidime-avibactam for treatment of severe KPC-producing Klebsiella pneumoniae infections: a retrospective comparative study. Clin Microbiol Infect. 2020;26(2):156-162.
Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin Infect Dis. 2016;62(10):e51-77.
MacDougall C, Polk RE. Antimicrobial stewardship programs in health care systems. Clin Microbiol Rev. 2005;18(4):638-656.
Pugh R, Grant C, Cooke RP, Dempsey G. Short-course versus prolonged-course antibiotic therapy for hospital-acquired pneumonia in critically ill adults. Cochrane Database Syst Rev. 2015;(8):CD007577.
Hecker SJ, Reddy KR, Lomovskaya O, et al. Discovery of cyclic boronic acid QPX7728, an ultra-broad-spectrum inhibitor of serine and metallo β-lactamases. J Med Chem. 2020;63(14):7491-7507.
Durand-Réville TF, Guler S, Comita-Prevoir J, et al. ETX2514 is a broad-spectrum β-lactamase inhibitor for the treatment of drug-resistant Gram-negative bacteria including Acinetobacter baumannii. Nat Microbiol. 2017;2(11):17104.
Dickey SW, Cheung GYC, Otto M. Different drugs for bad bugs: antivirulence strategies in the age of antibiotic resistance. Nat Rev Drug Discov. 2017;16(7):457-471.
Torres VJ, Stauff DL, Pishchany G, et al. A Staphylococcus aureus regulatory system that responds to host heme and modulates virulence. Cell Host Microbe. 2007;1(2):109-119.
Huh AJ, Kwon YJ. "Nanoantibiotics": a new paradigm for treating infectious diseases using nanomaterials in the antibiotics resistant era. J Control Release. 2011;156(2):128-145.
Yang JH, Wright SN, Hamblin M, et al. A white-box machine learning approach for revealing antibiotic mechanisms of action. Cell. 2019;177(6):1649-1661.e9.
Stokes JM, Yang K, Swanson K, et al. A deep learning approach to antibiotic discovery. Cell. 2020;180(4):688-702.e13.
Crews KR, Hicks JK, Pui CH, Relling MV, Evans WE. Pharmacogenomics and individualized medicine: translating science into practice. Clin Pharmacol Ther. 2012;92(4):467-475.
Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med. 2016;8(1):39.
Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):e61-e111.
Paul M, Carrara E, Retamar P, 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(4):521-547.
Bassetti M, Righi E, Carnelutti A, et al. Multidrug-resistant Klebsiella pneumoniae: challenges for treatment, prevention and infection control. Expert Rev Anti Infect Ther. 2018;16(10):749-761.
Jenkins TC, Knepper BC, Sabel AL, et al. Decreased antibiotic utilization after implementation of a guideline for inpatient cellulitis and cutaneous abscess. Arch Intern Med. 2011;171(12):1072-1079.
Schuts EC, Hulscher MEJL, Mouton JW, et al. Current evidence on hospital antimicrobial stewardship objectives: a systematic review and meta-analysis. Lancet Infect Dis. 2016;16(7):847-856.
Rex JH, Talbot GH, Goldberger MJ, et al. Progress in the fight against multidrug-resistant bacteria 2005-2016: modern noninferiority trial designs enable antibiotic development in advance of epidemic bacterial resistance. Clin Infect Dis. 2017;65(1):141-146.
Powers JH, Evans SR, Kesselheim AS. Studying new antibiotics for multidrug resistant infections: are today's patients paying for tomorrow's evidence? BMJ. 2018;360:k587.
Rex JH, Eisenstein BI, Alder J, et al. A comprehensive regulatory framework to address the unmet need for new antibiotics. Lancet Infect Dis. 2013;13(3):269-275.
Theuretzbacher U, Gottwalt S, Beyer P, et al. Analysis of the clinical antibacterial and antifungal pipeline. Lancet Infect Dis. 2019;19(2):e40-e50.
Humphries RM, Ambler J, Mitchell SL, et al. CLSI methods development and standardization working group best practices for evaluation of antimicrobial susceptibility tests. J Clin Microbiol. 2018;56(4):e01934-17.
Doron S, Davidson LE. Antimicrobial stewardship. Mayo Clin Proc. 2011;86(11):1113-1123.
Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis. 2013;13(12):1057-1098.
Outterson K, Gopinathan U, Clift C, et al. Delinking payment for essential medicines: time for action. Lancet. 2018;392(10162):2194-2201.
No comments:
Post a Comment