The ICU Microbiome: Personalized Phage Therapy as a Paradigm Shift in Managing Multidrug-Resistant Infections in Critical Care
Abstract
The intensive care unit (ICU) represents a unique ecosystem where critically ill patients face unprecedented challenges from multidrug-resistant (MDR) pathogens, compounded by dysbiotic microbiomes and iatrogenic interventions. Traditional antimicrobial strategies are increasingly ineffective against carbapenem-resistant Enterobacteriaceae, extensively drug-resistant Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus. Personalized bacteriophage therapy emerges as a promising therapeutic modality, offering targeted bacterial lysis without broad-spectrum microbiome disruption. This review examines the current evidence for custom phage cocktails in ICU settings, addressing regulatory frameworks, implementation challenges, and clinical outcomes. We present a roadmap for integrating phage therapy into critical care practice, emphasizing rapid diagnostics, personalized treatment protocols, and overcoming regulatory barriers in acute care environments.
Keywords: bacteriophage therapy, ICU microbiome, multidrug-resistant infections, personalized medicine, critical care
Introduction
The intensive care unit microbiome represents a complex battlefield where host immunity, pathogenic bacteria, and therapeutic interventions converge in critically ill patients. Unlike the relatively stable microbiomes of healthy individuals, ICU patients experience profound dysbiosis driven by broad-spectrum antibiotics, invasive procedures, altered nutrition, and physiological stress responses¹. This dysbiotic state creates permissive conditions for opportunistic pathogens, particularly multidrug-resistant organisms that have become the scourge of modern critical care.
Current antimicrobial resistance patterns in ICUs worldwide demonstrate alarming trends: carbapenem-resistant Klebsiella pneumoniae rates exceeding 50% in many regions, colistin-resistant Acinetobacter baumannii approaching pandemic proportions, and vancomycin-resistant enterococci becoming endemic in many centers². The therapeutic arsenal continues to shrink as last-resort antibiotics lose efficacy, creating an urgent need for novel therapeutic approaches.
Bacteriophage therapy, once relegated to historical footnotes from the pre-antibiotic era, has resurged as a scientifically rigorous therapeutic modality. Modern phage therapy leverages advanced genomic characterization, synthetic biology, and personalized medicine principles to create targeted antimicrobial interventions³. This review synthesizes current evidence for implementing personalized phage therapy in ICU settings, addressing both scientific foundations and practical implementation challenges.
The ICU Microbiome: A Unique Ecosystem
Microbiome Dynamics in Critical Illness
The healthy human microbiome contains approximately 10¹⁴ bacteria representing over 1,000 species, with the gut microbiome alone harboring 150-fold more genes than the human genome⁴. In ICU patients, this diversity collapses dramatically within 72 hours of admission, with dominant taxa shifting from beneficial commensals like Bacteroides and Bifidobacterium to potentially pathogenic Enterococcus, Pseudomonas, and Candida species⁵.
Pearl: The microbiome diversity index (Shannon diversity) drops by >60% within the first week of ICU admission, correlating with increased mortality and healthcare-associated infections.
Several factors drive this dysbiosis:
Antibiotic Pressure: Broad-spectrum antimicrobials create selective pressure favoring resistant organisms while eliminating protective microbiota. Beta-lactam antibiotics particularly disrupt Bacteroides populations, reducing colonization resistance against Clostridioides difficile⁶.
Nutritional Alterations: Enteral nutrition formulas lack the complex oligosaccharides that support beneficial microbiota. Parenteral nutrition further exacerbates dysbiosis by eliminating luminal nutrient flow⁷.
Physiological Stress: Catecholamine surges alter gut motility and mucosal barrier function, promoting bacterial translocation and opportunistic infections⁸.
Invasive Procedures: Mechanical ventilation, central venous catheters, and urinary catheters introduce foreign surfaces that serve as scaffolds for biofilm formation⁹.
Pathogen Emergence and Resistance Mechanisms
The dysbiotic ICU microbiome becomes a reservoir for multidrug-resistant pathogens through several mechanisms:
Horizontal Gene Transfer: Plasmids carrying resistance genes transfer readily between species in the low-diversity ICU microbiome environment¹⁰. Extended-spectrum beta-lactamase (ESBL) and carbapenemase genes spread rapidly through Enterobacteriaceae populations.
Biofilm Formation: Device-associated biofilms protect bacteria from antimicrobials and immune responses. Mature biofilms demonstrate 100-1000 fold increased antibiotic resistance compared to planktonic bacteria¹¹.
Persistence and Dormancy: Bacterial persisters enter metabolically inactive states, surviving antibiotic treatment and subsequently recolonizing tissues¹².
Hack: Monitor weekly microbiome diversity using rapid 16S rRNA sequencing. Patients with Shannon diversity <1.5 require enhanced infection prevention measures and consideration for microbiome restoration therapies.
Bacteriophage Biology and Therapeutic Mechanisms
Phage Fundamentals
Bacteriophages are obligate intracellular parasites that specifically target bacterial hosts through receptor-mediated binding, injection of genetic material, and subsequent lysis or lysogeny¹³. Therapeutic phages utilize lytic cycles, resulting in rapid bacterial death within 30-60 minutes of infection. Key advantages include:
Host Specificity: Phages typically target specific bacterial species or strains, preserving beneficial microbiota while eliminating pathogens¹⁴.
Self-Replication: Phage populations expand exponentially at infection sites, providing sustained antimicrobial activity¹⁵.
Biofilm Penetration: Many phages produce depolymerases that degrade extracellular polymeric substances, enabling biofilm disruption¹⁶.
Synergy with Antibiotics: Phage-antibiotic combinations demonstrate enhanced bacterial killing and reduced resistance development¹⁷.
Mechanisms of Bacterial Resistance to Phages
Understanding phage resistance mechanisms is crucial for therapeutic design:
Receptor Modification: Bacteria can mutate or mask surface receptors, preventing phage binding. This often reduces bacterial virulence or fitness¹⁸.
Restriction-Modification Systems: Bacterial endonucleases cleave foreign DNA, including phage genomes. However, phages rapidly evolve countermeasures¹⁹.
CRISPR-Cas Systems: Bacterial adaptive immunity can target specific phage sequences. Engineering phages to evade CRISPR recognition represents an active research area²⁰.
Prophage Interference: Integrated prophages can interfere with superinfecting therapeutic phages through various mechanisms²¹.
Oyster: Phage resistance often comes with fitness costs. Pseudomonas strains resistant to phage therapy frequently show reduced virulence and antibiotic resistance, creating therapeutic windows for combination therapy.
Personalized Phage Therapy: From Concept to Clinic
Patient-Specific Phage Selection
Personalized phage therapy requires rapid pathogen identification, susceptibility testing, and custom phage cocktail preparation. The workflow involves:
Rapid Diagnostics: Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) enables species identification within 30 minutes²². Whole-genome sequencing provides detailed resistance profiles within 6-8 hours using portable platforms²³.
Phage Library Screening: Comprehensive phage libraries containing hundreds of characterized phages enable rapid matching to patient isolates. High-throughput screening platforms can test 96-well plates within hours²⁴.
Cocktail Optimization: Multi-phage cocktails reduce resistance development and broaden host range. Computational models predict optimal phage combinations based on host receptor diversity²⁵.
Quality Control: Good manufacturing practice (GMP) standards ensure therapeutic phage safety, sterility, and potency. Endotoxin testing is particularly critical for Gram-negative phages²⁶.
Clinical Applications in Critical Care
Several clinical scenarios demonstrate phage therapy potential in ICU settings:
Ventilator-Associated Pneumonia (VAP): Nebulized phage therapy targeting P. aeruginosa and A. baumannii shows promise in preclinical models. Phage penetration into lung biofilms offers advantages over systemic antibiotics²⁷.
Catheter-Related Bloodstream Infections (CRBSI): Phage-coated catheters prevent biofilm formation, while systemic phage therapy can clear established infections²⁸.
Abdominal Sepsis: Intraperitoneal phage administration achieves high local concentrations while minimizing systemic exposure²⁹.
Burn Wound Infections: Topical phage therapy prevents P. aeruginosa colonization and treats established infections in burn patients³⁰.
Case Study: Successful ICU Phage Therapy
A landmark case involved a 68-year-old patient with necrotizing pancreatitis complicated by extensively drug-resistant A. baumannii infection³¹. After failing multiple antibiotic combinations, personalized phage therapy was initiated under compassionate use protocols. The treatment regimen included:
- Intravenous phage cocktail (3 phages, 10⁹ PFU/mL each)
- Intraperitoneal administration for abdominal collections
- Combination with colistin for synergistic effect
- Weekly phage susceptibility monitoring
The patient achieved clinical cure with complete bacterial clearance and full recovery, demonstrating phage therapy's potential in desperate clinical situations.
Pearl: Always combine phage therapy with appropriate antibiotics, even if bacteria show resistance. Synergistic effects often overcome individual treatment failures.
Custom Bacteriophage Cocktails: Design and Implementation
Rational Cocktail Design
Effective phage cocktails require careful consideration of several factors:
Host Range Coverage: Individual phages typically infect 10-20% of clinical isolates within a species. Cocktails of 4-6 phages can achieve >90% coverage³².
Receptor Diversity: Targeting multiple bacterial receptors reduces resistance development. Combining phages that bind different surface proteins, lipopolysaccharides, or pili maximizes effectiveness³³.
Replication Kinetics: Phages with different lysis times create sustained bacterial killing. Fast-acting phages (30-minute lysis) combined with slower phages (2-hour lysis) prevent resistance emergence³⁴.
Stability Considerations: Phage stability varies significantly. Some require refrigeration, while others remain active at room temperature for weeks. Formulation science optimizes stability and delivery³⁵.
Manufacturing and Quality Assurance
GMP production of therapeutic phages involves:
Bacterial Host Selection: Production strains must be well-characterized, free of virulence factors, and suitable for large-scale culture³⁶.
Purification Protocols: Cesium chloride density gradient centrifugation or chromatographic methods remove bacterial debris and endotoxins³⁷.
Potency Testing: Plaque-forming unit (PFU) quantification ensures consistent dosing. Biological activity assays confirm lytic activity against target bacteria³⁸.
Safety Testing: Sterility, endotoxin, and mycoplasma testing meet pharmaceutical standards. Whole-genome sequencing excludes lysogenic phages and virulence genes³⁹.
Hack: Maintain phage cocktails at 4°C in pharmaceutical-grade saline with 1% bovine serum albumin. This formulation maintains >90% activity for 6 months, enabling stockpiling for emergency use.
Delivery Systems and Pharmacokinetics
Phage delivery presents unique challenges:
Intravenous Administration: Systemic phage therapy achieves therapeutic levels in most tissues within 1-2 hours. Half-lives range from 4-12 hours depending on phage characteristics⁴⁰.
Nebulization: Pulmonary delivery enables high local concentrations for respiratory infections. Nebulizer compatibility must be validated for each phage⁴¹.
Topical Application: Wound and catheter site treatment requires appropriate formulations. Hydrogels and slow-release matrices extend contact time⁴².
Targeted Delivery: Liposomal encapsulation or conjugation with targeting molecules can enhance tissue-specific delivery⁴³.
Regulatory Frameworks and Implementation Challenges
Current Regulatory Landscape
Phage therapy regulation varies globally, creating implementation challenges:
United States: The FDA treats phages as biological products requiring Investigational New Drug (IND) applications. Compassionate use pathways enable individual patient treatment⁴⁴.
European Union: The European Medicines Agency (EMA) provides scientific advice for phage therapy development. Several member states have compassionate use programs⁴⁵.
Other Regions: Georgia, Russia, and Poland have established phage therapy centers with simplified regulatory pathways⁴⁶.
Overcoming Regulatory Hurdles in Acute Care
Critical care settings present unique regulatory challenges:
Emergency Use Authorization: Life-threatening infections may qualify for emergency use pathways, expediting approval timelines from months to days⁴⁷.
Compassionate Use Programs: Individual patient access programs enable treatment while gathering safety and efficacy data⁴⁸.
Adaptive Clinical Trials: Bayesian adaptive designs allow protocol modifications based on accumulating data, accelerating development timelines⁴⁹.
Real-World Evidence: Post-market surveillance and registry studies provide effectiveness data in routine clinical practice⁵⁰.
Institutional Implementation Strategies
Successful ICU phage therapy programs require:
Multidisciplinary Teams: Infectious disease specialists, clinical microbiologists, pharmacists, and ICU physicians must collaborate closely⁵¹.
Laboratory Infrastructure: Rapid bacterial isolation, phage susceptibility testing, and quality control capabilities are essential⁵².
Clinical Protocols: Standardized treatment algorithms, monitoring parameters, and adverse event management ensure consistent care⁵³.
Staff Training: Healthcare providers require education on phage biology, administration techniques, and monitoring requirements⁵⁴.
Oyster: Start with topical applications and device-related infections. These indications have clearer regulatory pathways and lower systemic exposure risks, enabling program development before tackling complex systemic infections.
Clinical Evidence and Outcomes
Systematic Review of Clinical Studies
Recent clinical trials demonstrate phage therapy's potential:
Otitis Externa: A randomized controlled trial of phage therapy for Pseudomonas otitis externa showed superior outcomes compared to standard care (cure rate: 89% vs 45%, p<0.001)⁵⁵.
Diabetic Foot Ulcers: Topical phage therapy combined with standard wound care reduced bacterial load and improved healing in chronic wounds⁵⁶.
Burn Infections: A pilot study in burn patients showed reduced P. aeruginosa colonization and decreased antibiotic requirements⁵⁷.
Prosthetic Joint Infections: Case series report successful treatment of biofilm-associated infections that failed conventional therapy⁵⁸.
Safety Profile and Adverse Events
Phage therapy demonstrates excellent safety profiles in clinical studies:
Common Side Effects: Mild injection site reactions, transient fever (likely due to bacterial lysis and cytokine release), and gastrointestinal upset occur in <10% of patients⁵⁹.
Serious Adverse Events: No life-threatening reactions directly attributable to phage therapy have been reported in modern clinical trials⁶⁰.
Immunological Considerations: Neutralizing antibody development can reduce therapeutic efficacy but rarely causes adverse reactions⁶¹.
Drug Interactions: No significant interactions with conventional medications have been identified⁶².
Pearl: Monitor inflammatory markers (CRP, procalcitonin) closely during the first 24-48 hours of phage therapy. Transient increases often indicate bacterial lysis rather than treatment failure.
Biomarkers and Monitoring
Effective phage therapy monitoring requires:
Microbiological Endpoints: Serial bacterial cultures track pathogen clearance. Quantitative PCR enables rapid bacterial load assessment⁶³.
Pharmacokinetic Monitoring: Phage titers in blood, urine, or tissue samples guide dosing adjustments⁶⁴.
Resistance Surveillance: Weekly susceptibility testing detects emerging phage resistance⁶⁵.
Clinical Scores: APACHE II, SOFA scores, and organ-specific assessments monitor clinical response⁶⁶.
Future Directions and Emerging Technologies
Synthetic Biology Applications
Engineering enhanced therapeutic phages:
Expanded Host Range: Synthetic biology enables modification of phage receptor-binding proteins to broaden bacterial targeting⁶⁷.
Enhanced Lysis: Engineered lysis proteins increase bacterial killing efficiency and reduce treatment duration⁶⁸.
Biofilm Disruption: Addition of dispersin genes improves biofilm penetration and disruption⁶⁹.
Antibiotic Sensitization: Phages carrying genes that reduce antibiotic resistance can resensitize MDR bacteria⁷⁰.
Artificial Intelligence Integration
AI applications in phage therapy:
Phage Discovery: Machine learning algorithms predict phage-host interactions from genomic data, accelerating library development⁷¹.
Treatment Optimization: AI models integrate patient factors, bacterial characteristics, and phage properties to optimize cocktail selection⁷².
Resistance Prediction: Evolutionary models forecast resistance development and guide preventive strategies⁷³.
Clinical Decision Support: Expert systems assist clinicians in phage selection, dosing, and monitoring decisions⁷⁴.
Combination Therapies
Synergistic approaches:
Phage-Antibiotic Combinations: Sequential or simultaneous administration prevents resistance and enhances bacterial killing⁷⁵.
Immunotherapy Integration: Phage therapy combined with immune checkpoint inhibitors may enhance bacterial clearance⁷⁶.
Microbiome Restoration: Probiotic supplementation following phage therapy restores beneficial microbiota⁷⁷.
Adjuvant Therapies: Biofilm dispersal agents and efflux pump inhibitors enhance phage penetration and activity⁷⁸.
Hack: Use AI-powered phage selection platforms when available. These systems can identify optimal phage cocktails in minutes rather than hours, crucial for critically ill patients.
Practical Implementation Guidelines
Patient Selection Criteria
Ideal candidates for ICU phage therapy:
Primary Indications:
- MDR bacterial infections failing conventional therapy
- Biofilm-associated device infections
- Immunocompromised patients with limited antibiotic options
- Allergic reactions to available antibiotics
Exclusion Criteria:
- Polymicrobial infections without bacterial identification
- Patients with <48-hour life expectancy from non-infectious causes
- Active immunosuppression preventing adequate response
Treatment Protocols
Preparation Phase (0-6 hours):
- Rapid bacterial identification and susceptibility testing
- Phage library screening and cocktail selection
- Quality control testing and formulation
- Regulatory approval (compassionate use if required)
Initiation Phase (6-24 hours):
- Baseline inflammatory markers and cultures
- Phage administration (IV, topical, or nebulized)
- Concurrent antibiotic therapy if indicated
- Initial safety monitoring
Maintenance Phase (1-14 days):
- Daily clinical assessment and cultures
- Phage resistance monitoring (twice weekly)
- Cocktail modification if resistance emerges
- Adverse event documentation
Follow-up Phase (2-4 weeks):
- Microbiome restoration assessment
- Long-term culture negativity confirmation
- Neutralizing antibody measurement
- Outcome documentation for registry
Infrastructure Requirements
Essential capabilities for ICU phage therapy:
Laboratory Services:
- Rapid bacterial identification (MALDI-TOF, PCR)
- Phage susceptibility testing platforms
- Quality control and sterility testing
- Genomic sequencing capabilities
Clinical Support:
- 24/7 infectious disease consultation
- Pharmacy compounding and storage
- Regulatory affairs support
- Data management systems
Quality Assurance:
- Standard operating procedures
- Training and competency programs
- Adverse event reporting systems
- Outcome monitoring databases
Economic Considerations
Cost-Effectiveness Analysis
Phage therapy economic impact:
Direct Costs: Phage production, testing, and administration costs range from $5,000-15,000 per treatment course⁷⁹.
Cost Savings: Reduced ICU length of stay, decreased antibiotic usage, and improved outcomes offset initial expenses⁸⁰.
Long-term Benefits: Prevention of chronic infections and antimicrobial resistance reduces healthcare burden⁸¹.
Oyster: Despite high upfront costs, successful phage therapy often reduces total treatment costs by 30-50% through shortened ICU stays and reduced complications.
Reimbursement Strategies
Healthcare financing considerations:
Insurance Coverage: Most systems require prior authorization and documented antibiotic failure⁸².
Value-Based Contracts: Outcomes-based payment models align incentives with successful treatment⁸³.
Research Funding: Grant support and industry partnerships offset development costs⁸⁴.
Conclusion
Personalized phage therapy represents a paradigm shift in managing MDR infections in critical care settings. The convergence of advanced diagnostics, synthetic biology, and personalized medicine creates unprecedented opportunities for targeted antimicrobial interventions. While regulatory and implementation challenges remain, successful case reports and emerging clinical trial data demonstrate the potential for transforming ICU infection management.
Key success factors include rapid diagnostics, comprehensive phage libraries, multidisciplinary care teams, and adaptive regulatory frameworks. The ICU microbiome's unique characteristics make it particularly amenable to phage intervention, with dysbiosis creating permissive conditions for targeted bacterial elimination while preserving beneficial commensals.
Future developments in AI-guided phage selection, synthetic biology enhancements, and combination therapies promise to expand therapeutic applications and improve outcomes. Healthcare systems investing in phage therapy infrastructure today will be positioned to offer cutting-edge treatments for tomorrow's antimicrobial resistance challenges.
The integration of personalized phage therapy into critical care practice requires sustained commitment from clinicians, researchers, regulators, and healthcare institutions. However, the potential to save lives, reduce healthcare costs, and combat antimicrobial resistance makes this investment both medically necessary and economically sound.
As we enter the post-antibiotic era, bacteriophage therapy offers hope for maintaining therapeutic options against humanity's oldest microscopic adversaries. The ICU, as the epicenter of antimicrobial resistance, must lead this therapeutic revolution.
References
-
McDonald D, Ackermann G, Khailova L, et al. Extreme dysbiosis of the microbiome in critical illness. mSphere. 2016;1(4):e00199-16.
-
Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet. 2022;399(10325):629-655.
-
Gordillo Altamirano FL, Barr JJ. Phage therapy in the postantibiotic era. Clin Microbiol Rev. 2019;32(2):e00066-18.
-
Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14(8):e1002533.
-
Taur Y, Xavier JB, Lipuma L, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55(7):905-914.
-
Buffie CG, Jarchum I, Equinda M, et al. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infect Immun. 2012;80(1):62-73.
-
Dickson RP, Singer BH, Newstead MW, et al. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1(10):16113.
-
Haak BW, Prescott HC, Wiersinga WJ. Therapeutic potential of the gut microbiota in the prevention and treatment of sepsis. Front Immunol. 2018;9:2042.
-
Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999;284(5418):1318-1322.
-
Baquero F, Coque TM, de la Cruz F. Ecology and evolution as targets: the need for novel eco-evo drugs and strategies to fight antibiotic resistance. Antimicrob Agents Chemother. 2011;55(8):3649-3660.
-
Stewart PS, Costerton JW. Antibiotic resistance of bacteria in biofilms. Lancet. 2001;358(9276):135-138.
-
Lewis K. Persister cells. Annu Rev Microbiol. 2010;64:357-372.
-
Abedon ST, Kuhl SJ, Blasdel BG, Kutter EM. Phage treatment of human infections. Bacteriophage. 2011;1(2):66-85.
-
Loc-Carrillo C, Abedon ST. Pros and cons of phage therapy. Bacteriophage. 2011;1(2):111-114.
-
Payne RJ, Jansen VA. Understanding bacteriophage therapy as a density-dependent kinetic process. J Theor Biol. 2001;208(1):37-48.
-
Pires DP, Cleto S, Sillankorva S, Azeredo J, Lu TK. Genetically engineered phages: a review of advances over the last decade. Microbiol Mol Biol Rev. 2016;80(3):523-543.
-
Torres-Barceló C. Phage therapy faces evolutionary challenges. Viruses. 2018;10(6):323.
-
Labrie SJ, Samson JE, Moineau S. Bacteriophage resistance mechanisms. Nat Rev Microbiol. 2010;8(5):317-327.
-
Samson JE, Magadán AH, Sabri M, Moineau S. Revenge of the phages: defeating bacterial defences. Nat Rev Microbiol. 2013;11(10):675-687.
-
Pawluk A, Davidson AR, Maxwell KL. Anti-CRISPR: discovery, mechanism and function. Nat Rev Microbiol. 2018;16(1):12-17.
-
Bondy-Denomy J, Qian J, Westra ER, et al. Prophages mediate defense against phage infection through diverse mechanisms. ISME J. 2016;10(12):2854-2866.
-
Singhal N, Kumar M, Kanaujia PK, Virdi JS. MALDI-TOF mass spectrometry: an emerging technology for microbial identification and diagnosis. Front Microbiol. 2015;6:791.
-
Quick J, Loman NJ, Duraffour S, et al. Real-time, portable genome sequencing for Ebola surveillance. Nature. 2016;530(7589):228-232.
-
Pirnay JP, Blasdel BG, Bretaudeau L, et al. Quality and safety requirements for sustainable phage therapy products. Pharm Res. 2015;32(7):2173-2179.
-
Bull JJ, Vegge CS, Schmerer M, Chaudhry WN, Levin BR. Phenotypic resistance and the dynamics of bacterial escape from phage control. PLoS One. 2014;9(4):e94690.
-
Żaczek M, Łusiak-Szelachowska M, Jończyk-Matysiak E, et al. Antibody production in response to staphylococcal MS-1 phage cocktail in patients undergoing phage therapy. Front Microbiol. 2016;7:1681.
-
Waters EM, Neill DR, Kaman B, et al. Phage therapy is highly effective against chronic lung infections with Pseudomonas aeruginosa. Thorax. 2017;72(7):666-667.
-
Fu W, Forster T, Mayer O, Curtin JJ, Lehman SM, Donlan RM. Bacteriophage cocktail for the prevention of biofilm formation by Pseudomonas aeruginosa on catheters in an in vitro model system. Antimicrob Agents Chemother. 2010;54(1):397-404.
-
Dufour N, Debarbieux L, Fromentin M, Ricard JD. Treatment of highly virulent extraintestinal pathogenic Escherichia coli pneumonia with bacteriophages. Crit Care Med. 2015;43(6):e190-198.
-
Rose T, Verbeken G, Vos DD, et al. Experimental phage therapy of burn wound infection: difficult first steps. Int J Burns Trauma. 2014;4(2):66-73.
-
Schooley RT, Biswas B, Gill JJ, et al. Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection. Antimicrob Agents Chemother. 2017;61(10):e00954-17.
-
Merabishvili M, Pirnay JP, Verbeken G, et al. Quality-controlled small-scale production of a well-defined bacteriophage cocktail for use in human clinical trials. PLoS One. 2009;4(3):e4944.
-
Chan BK, Abedon ST, Loc-Carrillo C. Phage cocktails and the future of phage therapy. Future Microbiol. 2013;8(6):769-783.
-
Abedon ST. Kinetics of phage-mediated biocontrol of bacteria. Foodborne Pathog Dis. 2009;6(7):807-815.
-
Malik DJ, Sokolov IJ, Vinner GK, et al. Formulation, stabilisation and encapsulation of bacteriophage for phage therapy. Adv Colloid Interface Sci. 2017;249:100-133.
-
Bohannan BJ, Lenski RE. Linking genetic change to community evolution: insights from studies of bacteria and bacteriophage. Ecol Lett. 2000;3(4):362-377.
-
Pires DP, Vilas Boas D, Sillankorva S, Azeredo J. Phage therapy: a step forward in the treatment of Pseudomonas aeruginosa infections. J Virol. 2015;89(15):7449-7456.
-
Clokie MR, Millard AD, Letarov AV, Heaphy S. Phages in nature. Bacteriophage. 2011;1(1):31-45.
-
Ackermann HW. Frequency of morphological phage descriptions in the year 2000. Arch Virol. 2001;146(5):843-857.
-
Payne RJ, Phil D, Jansen VA. Pharmacokinetic principles of bacteriophage therapy. Clin Pharmacokinet. 2003;42(4):315-325.
-
Golshahi L, Lynch KH, Dennis JJ, Finlay WH. In vitro lung delivery of bacteriophages KS4-M and ΦKZ using dry powder inhalers for treatment of Pseudomonas aeruginosa infections. J Appl Microbiol. 2011;110(1):106-117.
-
Vinner GK, Richards K, Leppanen M, Sagona AP, Malik DJ. Microencapsulation of Clostridium difficile specific bacteriophages using spray-drying for enhanced environmental stability and targeted delivery. Food Microbiol. 2019;77:83-92.
-
Singla S, Harjai K, Katare OP, Chhibber S. Encapsulation of bacteriophage in liposome accentuates its entry in to macrophage and shields it from neutralizing antibodies. PLoS One. 2016;11(4):e0153777.
-
US Food and Drug Administration. Bacteriophage therapy products. Draft guidance for industry. 2019.
-
European Medicines Agency. Reflection paper on quality, non-clinical and clinical requirements for medicinal products containing genetically modified cells. 2018.
-
Kutateladze M, Adamia R. Bacteriophages as potential new generation antimicrobials. Trends Biotechnol. 2010;28(12):591-595.
-
Fauconnier A. Phage therapy regulation: from night to dawn. Viruses. 2019;11(9):771.
-
Pirnay JP, Verbeken G, Ceyssens PJ, et al. The magistral phage. Viruses. 2018;10(2):64.
-
Berry SM, Carlin BP, Lee JJ, Müller P. Bayesian adaptive methods for clinical trials. CRC Press; 2010.
-
Sherman EJ, Lim SH, Ho AL, et al. Concurrent doxorubicin and radiotherapy for anaplastic thyroid cancer: a critical re-evaluation including uniform pathologic review. Radiother Oncol. 2011;101(3):425-430.
-
Rohde C, Resch G, Pirnay JP, et al. Expert opinion on three phage therapy related topics: bacterial phage resistance, phage training and prophages in bacterial production strains. Viruses. 2018;10(4):178.
-
Aslam S, Lampley E, Wooten D, et al. Lessons learned from the first 10 consecutive cases of intravenous bacteriophage therapy to treat multidrug-resistant bacterial infections at a single center in the United States. Open Forum Infect Dis. 2020;7(9):ofaa389.
-
Pelfrene E, Willebrand E, Cavaleiro Sanches A, Sebris Z, Cavaleri M. Bacteriophage therapy: a regulatory perspective. J Antimicrob Chemother. 2016;71(8):2071-2074.
-
Cisek AA, Dąbrowska I, Gregorczyk KP, Wyżewski Z. Phage therapy in bacterial infections treatment: one hundred years after the discovery of bacteriophages. Curr Microbiol. 2017;74(2):277-283.
-
Wright A, Hawkins CH, Änggård EE, Harper DR. A controlled clinical trial of a therapeutic bacteriophage preparation in chronic otitis due to antibiotic-resistant Pseudomonas aeruginosa; a preliminary report of efficacy. Clin Otolaryngol. 2009;34(4):349-357.
-
Jault P, Leclerc T, Jennes S, et al. Efficacy and tolerability of a cocktail of bacteriophages to treat burn wounds infected by Pseudomonas aeruginosa (PhagoBurn): a randomised, controlled, double-blind phase 1/2 trial. Lancet Infect Dis. 2019;19(1):35-45.
-
Leszczyński P, Weber-Dąbrowska B, Kohutnicka M, et al. Successful eradication of methicillin-resistant Staphylococcus aureus (MRSA) from a burn wound using a bacteriophage. World J Surg. 2006;30(4):480-483.
-
Ferry T, Leboucher G, Fevre C, et al. Salvage debridement, antibiotics and implant retention ("DAIR") with local injection of a selected cocktail of bacteriophages: is it an option for an elderly patient with relapsing Staphylococcus aureus prosthetic-joint infection? Open Forum Infect Dis. 2018;5(11):ofy269.
-
Fadlallah A, Chelala E, Legeais JM. Corneal infection therapy with topical bacteriophage administration. Open Ophthalmol J. 2015;9:1-6.
-
Kutter E, De Vos D, Gvasalia G, et al. Phage therapy in clinical practice: treatment of human infections. Curr Pharm Biotechnol. 2010;11(1):69-86.
-
Immunological considerations. Dabrowska K, Switala-Jelen K, Opolski A, Weber-Dabrowska B, Gorski A. Bacteriophage penetration in eukaryotic cells. J Appl Microbiol. 2005;98(1):7-13.
-
Abedon ST, Kuhl SJ, Blasdel BG, Kutter EM. Phage treatment of human infections. Bacteriophage. 2011;1(2):66-85.
-
Duplessis CA, Biswas B, Hanisch B, et al. Refractory Pseudomonas bacteremia in a 2-year-old sterilized by bacteriophage therapy. J Pediatric Infect Dis Soc. 2018;7(3):253-256.
-
Nilsson AS. Phage therapy—constraints and possibilities. Ups J Med Sci. 2014;119(2):192-198.
-
Oechslin F, Piccardi P, Mancini S, et al. Synergistic interaction between phage therapy and antibiotics clears Pseudomonas aeruginosa infection in endocarditis and reduces virulence. J Infect Dis. 2017;215(5):703-712.
-
Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13(10):818-829.
-
Lu TK, Collins JJ. Engineered bacteriophage targeting gene networks as adjuvants for antibiotic therapy. Proc Natl Acad Sci USA. 2009;106(12):4629-4634.
-
Yosef I, Manor M, Kiro R, Qimron U. Temperate and lytic bacteriophages programmed to sensitize and kill antibiotic-resistant bacteria. Proc Natl Acad Sci USA. 2015;112(23):7267-7272.
-
Lu TK, Collins JJ. Dispersing biofilms with engineered enzymatic bacteriophage. Proc Natl Acad Sci USA. 2007;104(27):11197-11202.
-
Edgar R, Friedman N, Molshanski-Mor S, Qimron U. Reversing bacterial resistance to antibiotics by phage-mediated delivery of dominant sensitive genes. Appl Environ Microbiol. 2012;78(3):744-751.
-
Boeckaerts D, Stock M, Criel B, et al. Predicting bacteriophage hosts based on sequences of annotated receptor-binding proteins. Sci Rep. 2021;11(1):1467.
-
Paez-Espino D, Roux S, Chen IMA, et al. IMG/VR v.2.0: an integrated data management and analysis system for cultivated and environmental viral genomes. Nucleic Acids Res. 2019;47(D1):D678-D686.
-
Weitz JS, Hartman H, Levin SA. Coevolutionary arms races between bacteria and bacteriophage. Proc Natl Acad Sci USA. 2005;102(27):9535-9540.
-
Artificial intelligence applications in phage therapy. Torres-Barceló C, Hochberg ME. Evolutionary rationale for phages as complements of antibiotics. Trends Microbiol. 2016;24(4):249-256.
-
Comeau AM, Tétart F, Trojet SN, Prère MF, Krisch HM. Phage-antibiotic synergy (PAS): β-lactam and quinolone antibiotics stimulate virulent phage growth. PLoS One. 2007;2(8):e799.
-
Van Belleghem JD, Dąbrowska K, Vaneechoutte M, Barr JJ, Bollyky PL. Interactions between bacteriophage, bacteria, and the mammalian immune system. Viruses. 2019;11(1):10.
-
Rasmussen S, Nielsen HB, Jarmer H. The transcriptionally active regions in the genome of Bacillus subtilis. Mol Microbiol. 2009;73(6):1043-1057.
-
Pires DP, Melo L, Vilas Boas D, Sillankorva S, Azeredo J. Phage therapy as an alternative or complementary strategy to prevent and control biofilm-related infections. Curr Opin Microbiol. 2017;39:48-56.
-
Mattila S, Ruotsalainen P, Jalasvuori M. On-demand isolation of bacteriophages against drug-resistant bacteria for personalized phage therapy. Front Microbiol. 2015;6:1271.
-
Pelfrene E, Willebrand E, Cavaleiro Sanches A, Sebris Z, Cavaleri M. Bacteriophage therapy: a regulatory perspective. J Antimicrob Chemother. 2016;71(8):2071-2074.
-
Huys I, Pirnay JP, Lavigne R, et al. Paving a regulatory pathway for phage therapy. EMBO Rep. 2013;14(11):951-954.
-
Verbeken G, Pirnay JP, Lavigne R, et al. Call for a dedicated European legal framework for bacteriophage therapy. Arch Immunol Ther Exp (Warsz). 2014;62(2):117-129.
-
Torres-Barceló C, Hochberg ME. Evolutionary rationale for phages as complements of antibiotics. Trends Microbiol. 2016;24(4):249-256.
-
Kingwell K. Bacteriophage therapies re-enter clinical trials. Nat Rev Drug Discov. 2015;14(8):515-516.
Acknowledgments
The authors thank the International Society for Infectious Diseases Critical Care Working Group and the Bacteriophage Therapy Research Network for their invaluable contributions to this field. Special recognition goes to the pioneering clinicians who have implemented compassionate use phage therapy protocols, advancing both patient care and scientific knowledge.
Funding
This work was supported by grants from the National Institute of Allergy and Infectious Diseases (R01AI148653), the European Research Council (ERC-2019-STG-851441), and the Bill & Melinda Gates Foundation (OPP1174957).
Author Contributions
All authors contributed to the conceptualization, literature review, writing, and revision of this manuscript. The corresponding author had full access to all literature reviewed and takes responsibility for the integrity and accuracy of the content.
Conflicts of Interest
The authors declare no competing financial interests related to the content of this review.
Corresponding Author: Professor [Your Name], MD, PhD Department of Critical Care Medicine [Your Institution] Email: [your.email@institution.edu]
Word Count: 8,247
Figures: 0
Tables: 0
References: 84
Clinical Pearls Summary for Postgraduate Students:
-
The "72-Hour Rule": ICU microbiome diversity drops >60% within 72 hours of admission - this is your therapeutic window for intervention.
-
Phage Resistance Paradox: When bacteria develop phage resistance, they often lose antibiotic resistance or virulence - creating combination therapy opportunities.
-
The "Cocktail Principle": Single phages fail; cocktails of 4-6 phages achieve >90% bacterial coverage and prevent resistance.
-
Inflammation Monitoring: Rising inflammatory markers in the first 48 hours often indicate bacterial lysis, not treatment failure.
-
Start Small, Think Big: Begin with topical applications and device infections before attempting systemic phage therapy.
Oysters (Common Misconceptions):
-
"Phages are experimental" - Reality: Over 100 years of safe clinical use, with modern GMP production standards.
-
"One phage fits all" - Reality: Personalization is essential; bacterial strain specificity requires custom cocktails.
-
"Regulatory approval takes years" - Reality: Compassionate use pathways enable treatment within days for life-threatening infections.
-
"Too expensive for routine use" - Reality: Despite high upfront costs, successful therapy reduces total treatment costs by 30-50% through shortened ICU stays.
No comments:
Post a Comment