Tuesday, June 3, 2025

 

Step-by-Step Management of Parkinson's Disease: A Comprehensive Review

Dr Neeraj Manikath, Claude.ai

Abstract

Background: Parkinson's disease (PD) is the second most common neurodegenerative disorder, affecting over 10 million individuals worldwide. Optimal management requires a systematic, multidisciplinary approach that evolves with disease progression.

Objective: To provide a comprehensive, evidence-based framework for the step-by-step management of Parkinson's disease from diagnosis through advanced stages.

Methods: This review synthesizes current evidence from major clinical trials, international guidelines, and recent meta-analyses to present a structured approach to PD management.

Results: We present a systematic management framework encompassing: (1) accurate diagnosis and differential diagnosis, (2) initial therapeutic decisions, (3) optimization of dopaminergic therapy, (4) management of motor complications, (5) non-motor symptom recognition and treatment, (6) advanced therapies, and (7) palliative and end-of-life care considerations.

Conclusions: Effective PD management requires individualized, stepwise therapeutic escalation combined with comprehensive non-motor symptom management and timely consideration of advanced therapies to optimize quality of life throughout the disease course.

Keywords: Parkinson's disease, movement disorders, dopamine, levodopa, deep brain stimulation, motor complications


Introduction

Parkinson's disease represents a complex neurodegenerative condition characterized by progressive loss of dopaminergic neurons in the substantia nigra, resulting in the cardinal motor features of bradykinesia, rigidity, tremor, and postural instability¹. Beyond motor manifestations, PD encompasses a broad spectrum of non-motor symptoms that significantly impact quality of life and often precede motor symptoms by years².

The management of PD has evolved considerably with advances in understanding disease pathophysiology, expanded therapeutic options, and recognition of the importance of individualized care. This review provides a systematic, step-by-step approach to PD management based on current evidence and international consensus guidelines³⁻⁵.


Step 1: Accurate Diagnosis and Assessment

Clinical Diagnosis

The diagnosis of PD remains clinical, based on the presence of bradykinesia plus at least one of: muscular rigidity, rest tremor (4-6 Hz), or postural instability not caused by primary visual, vestibular, cerebellar, or proprioceptive dysfunction⁶.

Essential Diagnostic Criteria:

  • Bradykinesia (slowness of movement with progressive reduction in amplitude/speed)
  • Plus one of: muscular rigidity, rest tremor, postural instability

Supportive Criteria:

  • Unilateral onset with persistent asymmetry
  • Excellent response to levodopa (>70% improvement)
  • Severe levodopa-induced dyskinesia
  • Levodopa response ≥5 years
  • Clinical course ≥10 years

Differential Diagnosis

Careful exclusion of alternative diagnoses is crucial, particularly atypical parkinsonism:

Progressive Supranuclear Palsy (PSP):

  • Early falls, supranuclear gaze palsy, axial rigidity
  • Poor levodopa response

Multiple System Atrophy (MSA):

  • Autonomic failure, cerebellar signs, pyramidal signs
  • Stridor, poor levodopa response

Corticobasal Degeneration (CBD):

  • Asymmetric rigidity, apraxia, alien limb phenomenon
  • Cortical sensory loss

Lewy Body Dementia:

  • Early cognitive impairment, visual hallucinations
  • Fluctuating cognition, REM sleep behavior disorder

Baseline Assessment

Motor Assessment:

  • Unified Parkinson's Disease Rating Scale (MDS-UPDRS) Parts I-IV⁷
  • Hoehn and Yahr staging
  • Timed motor tasks (finger tapping, rapid alternating movements)

Non-Motor Assessment:

  • Cognitive screening (Montreal Cognitive Assessment)
  • Mood assessment (Beck Depression Inventory)
  • Sleep evaluation (Epworth Sleepiness Scale)
  • Autonomic function assessment
  • Quality of life measures (PDQ-39)

Diagnostic Imaging:

  • DaTscan (dopamine transporter SPECT) when diagnosis uncertain⁸
  • MRI to exclude structural lesions
  • Consider cardiac MIBG scintigraphy for differential diagnosis

Step 2: Initial Therapeutic Decisions

Treatment Initiation Timing

Treatment initiation should be individualized based on:

  • Functional impairment and quality of life impact
  • Occupational requirements
  • Patient age and comorbidities
  • Patient preferences regarding potential side effects

Key Principle: Treatment should begin when symptoms interfere with daily activities or quality of life, not merely upon diagnosis⁹.

First-Line Therapeutic Options

For Patients <65 Years

Preferred Initial Therapies:

  1. Dopamine Agonists (ropinirole, pramipexole, rotigotine)

    • Lower risk of motor complications
    • Gradual titration required
    • Monitor for impulse control disorders
  2. MAO-B Inhibitors (rasagiline, selegiline, safinamide)

    • Mild symptomatic benefit
    • Potential neuroprotective effects
    • Good tolerability profile

For Patients ≥65 Years

Preferred Initial Therapy:

  1. Levodopa/Carbidopa
    • Most effective symptomatic therapy
    • Better tolerability in elderly
    • Lower risk of psychiatric side effects

Monotherapy vs. Combination Therapy

Initial Monotherapy Preferred:

  • Allows assessment of individual drug response
  • Simplifies side effect attribution
  • Facilitates dose optimization

Early Combination Considerations:

  • Inadequate monotherapy response
  • Tremor-predominant disease (consider anticholinergics in young patients)
  • Specific symptom targeting

Step 3: Optimization of Dopaminergic Therapy

Levodopa Optimization

Starting Doses:

  • Immediate-release levodopa/carbidopa: 25/100 mg TID
  • Titrate by 25/100 mg every 3-7 days
  • Target: minimum effective dose for symptom control

Extended-Release Formulations:

  • Consider for patients with wearing-off
  • Rytary (extended-release carbidopa/levodopa)
  • Requires dose conversion and timing adjustments

Dopamine Agonist Optimization

Ropinirole Titration:

  • Week 1: 0.25 mg TID
  • Increase by 0.25 mg TID weekly
  • Maximum: 8 mg TID

Pramipexole Titration:

  • Week 1: 0.125 mg TID
  • Increase by 0.125 mg TID weekly
  • Maximum: 1.5 mg TID

Rotigotine Patch:

  • Starting dose: 2 mg/24 hours
  • Increase by 2 mg weekly
  • Maximum: 8 mg/24 hours

Monitoring and Adjustment

Regular Assessment (Every 3-6 months):

  • Motor symptom control
  • Activities of daily living
  • Side effect monitoring
  • Quality of life measures

Dose Adjustment Principles:

  • Optimize before adding additional medications
  • Consider timing of doses relative to meals
  • Address individual symptom variability

Step 4: Management of Motor Complications

Wearing-Off Phenomenon

Recognition:

  • Return of parkinsonian symptoms before next dose
  • Predictable symptom fluctuations
  • Shortened duration of benefit

Management Strategies:

  1. Increase Dosing Frequency

    • Reduce dosing intervals
    • Maintain total daily dose initially
  2. Add COMT Inhibitors

    • Entacapone 200 mg with each levodopa dose
    • Prolongs levodopa half-life
    • Stalevo (carbidopa/levodopa/entacapone combination)
  3. Extended-Release Formulations

    • Rytary for smoother plasma levels
    • Inbrija (inhaled levodopa) for off episodes
  4. Adjunctive Therapies

    • MAO-B inhibitors (rasagiline, safinamide)
    • Dopamine agonists if not already prescribed

Dyskinesia Management

Peak-Dose Dyskinesia:

  • Reduce individual levodopa doses
  • Increase dosing frequency
  • Add amantadine 100-300 mg daily¹⁰

Diphasic Dyskinesia:

  • More complex pattern
  • May require continuous dopaminergic stimulation
  • Consider advanced therapies earlier

Off-Period Dystonia:

  • Often affects feet/toes in early morning
  • Extend overnight dopaminergic coverage
  • Consider controlled-release preparations

Advanced Motor Complications

Complex Fluctuations:

  • Unpredictable on-off phenomena
  • Delayed-on, no-on responses
  • Freezing episodes

Management Approach:

  • Optimize oral medications first
  • Consider advanced therapies (DBS, pump therapies)
  • Multidisciplinary team involvement

Step 5: Non-Motor Symptom Management

Cognitive Symptoms

Mild Cognitive Impairment:

  • Cognitive rehabilitation
  • Optimize dopaminergic medications
  • Address contributing factors (depression, sleep disorders)

Parkinson's Disease Dementia:

  • Rivastigmine 3-12 mg daily¹¹
  • Reduce anticholinergic burden
  • Behavioral interventions

Psychiatric Symptoms

Depression:

  • SSRIs (sertraline, citalopram) first-line
  • Consider tricyclic antidepressants
  • Pramipexole may have antidepressant effects

Anxiety:

  • Often responds to dopaminergic optimization
  • SSRIs for persistent anxiety
  • Cognitive-behavioral therapy

Psychosis:

  • Reduce dopaminergic medications if possible
  • Quetiapine 12.5-50 mg daily
  • Pimavanserin 34 mg daily (FDA-approved for PD psychosis)¹²

Sleep Disorders

REM Sleep Behavior Disorder:

  • Melatonin 3-12 mg at bedtime
  • Clonazepam 0.5-2 mg at bedtime
  • Bedroom safety measures

Excessive Daytime Sleepiness:

  • Modafinil 100-400 mg daily
  • Address sleep hygiene
  • Evaluate for sleep apnea

Restless Legs Syndrome:

  • Often responds to dopamine agonists
  • Iron supplementation if deficient
  • Gabapentin for refractory cases

Autonomic Symptoms

Orthostatic Hypotension:

  • Non-pharmacologic measures (hydration, compression stockings)
  • Fludrocortisone 0.1-0.3 mg daily
  • Midodrine 2.5-10 mg TID
  • Droxidopa 100-600 mg TID¹³

Constipation:

  • Increased fiber and fluid intake
  • Polyethylene glycol
  • Lubiprostone for refractory cases

Urinary Dysfunction:

  • Evaluate for retention vs. overactivity
  • Anticholinergics for overactive bladder
  • Alpha-blockers for retention

Speech and Swallowing

Hypophonia:

  • Lee Silverman Voice Treatment (LSVT LOUD)
  • Speech therapy referral
  • Consider voice amplification devices

Dysphagia:

  • Speech-language pathology evaluation
  • Modified barium swallow study
  • Dietary modifications
  • Consider PEG tube for severe cases

Step 6: Advanced Therapies

Deep Brain Stimulation (DBS)

Candidacy Criteria:

  • Good levodopa response (>30% improvement in UPDRS-III)
  • Motor complications despite optimal medical therapy
  • Age typically <70-75 years
  • Absence of significant cognitive impairment
  • Realistic expectations

Target Selection:

  • Subthalamic Nucleus (STN): Best for tremor, rigidity, bradykinesia
  • Globus Pallidus Internus (GPi): Preferred for dyskinesia-predominant patients

Expected Outcomes:

  • 30-60% improvement in motor symptoms
  • Significant reduction in motor complications
  • Medication reduction possible

Continuous Therapies

Duopa (Carbidopa/Levodopa Enteral Suspension):

  • Percutaneous gastrostomy administration
  • Continuous dopaminergic stimulation
  • Reduces motor fluctuations significantly¹⁴

Apomorphine Pump:

  • Continuous subcutaneous infusion
  • Rapid onset of action
  • Requires antiemetic pretreatment

Patient Selection for Advanced Therapies

Ideal Candidates:

  • Significant motor complications
  • Good cognitive function
  • Realistic expectations
  • Adequate social support
  • Failed optimal medical management

Relative Contraindications:

  • Significant cognitive impairment
  • Active psychiatric disease
  • Poor surgical candidate
  • Unrealistic expectations

Step 7: Multidisciplinary Care and Supportive Therapies

Physical Therapy

Goals:

  • Maintain mobility and flexibility
  • Improve balance and reduce falls
  • Address freezing episodes
  • Gait training

Specific Interventions:

  • Large amplitude movements (LSVT BIG)
  • Cueing strategies for freezing
  • Balance training programs
  • Strength and endurance exercises

Occupational Therapy

Focus Areas:

  • Activities of daily living
  • Home safety assessment
  • Adaptive equipment
  • Energy conservation techniques

Exercise Programs

Evidence-Based Benefits:

  • Forced exercise (high-intensity cycling)
  • Tango dancing
  • Tai Chi for balance
  • Boxing programs (Rock Steady Boxing)

General Recommendations:

  • 150 minutes moderate exercise weekly
  • Include aerobic, strength, and flexibility components
  • Balance training 2-3 times weekly

Nutritional Considerations

Protein Timing:

  • Separate protein intake from levodopa doses
  • Consider low-protein breakfast and lunch
  • Concentrate protein at dinner

Specific Nutrients:

  • Adequate calcium and vitamin D
  • B-vitamin supplementation if deficient
  • Maintain adequate fiber intake

Step 8: Monitoring and Long-Term Management

Regular Assessment Schedule

Every 3-6 Months:

  • Motor symptom evaluation (MDS-UPDRS)
  • Non-motor symptom screening
  • Medication review and optimization
  • Functional status assessment

Annual Assessments:

  • Comprehensive cognitive evaluation
  • Bone density screening
  • Cardiovascular risk assessment
  • Advanced therapy candidacy review

Disease Progression Monitoring

Early Disease (Hoehn & Yahr 1-2):

  • Focus on symptom control
  • Lifestyle modifications
  • Exercise programs
  • Education and support

Moderate Disease (Hoehn & Yahr 2.5-3):

  • Motor complication management
  • Non-motor symptom treatment
  • Advanced therapy consideration
  • Safety assessments

Advanced Disease (Hoehn & Yahr 4-5):

  • Palliative care consultation
  • Caregiver support
  • End-of-life planning
  • Comfort-focused care

Quality Indicators

Treatment Goals:

  • Optimize functional independence
  • Minimize motor complications
  • Address non-motor symptoms
  • Maintain quality of life
  • Prevent complications

Red Flags Requiring Urgent Review:

  • Sudden worsening of symptoms
  • New psychiatric symptoms
  • Falling episodes
  • Swallowing difficulties
  • Medication non-adherence

Special Populations and Considerations

Young-Onset Parkinson's Disease

Unique Considerations:

  • Higher risk of motor complications
  • Different psychological impact
  • Family planning considerations
  • Career implications

Management Modifications:

  • Delayed levodopa initiation when possible
  • Dopamine agonist preference
  • Early DBS consideration
  • Genetic counseling

Elderly Patients

Special Considerations:

  • Increased risk of psychiatric side effects
  • Polypharmacy interactions
  • Falls risk
  • Cognitive vulnerability

Management Approach:

  • Start low, go slow
  • Prefer levodopa over dopamine agonists
  • Careful monitoring for confusion
  • Fall prevention strategies

Comorbid Conditions

Cardiovascular Disease:

  • Monitor for orthostatic hypotension
  • Drug interaction awareness
  • Exercise program modifications

Diabetes:

  • Glucose control optimization
  • Neuropathy vs. PD symptom differentiation
  • Medication timing considerations

Emerging Therapies and Future Directions

Novel Therapeutic Targets

Alpha-Synuclein Targeting:

  • Immunotherapy approaches
  • Small molecule inhibitors
  • Gene therapy strategies

Neuroprotection:

  • GLP-1 receptor agonists
  • Antioxidant strategies
  • Mitochondrial therapies

Precision Medicine:

  • Genetic subtyping
  • Biomarker-guided therapy
  • Personalized treatment algorithms

Technology Integration

Digital Health Tools:

  • Smartphone-based symptom monitoring
  • Wearable device integration
  • Telemedicine platforms
  • AI-assisted clinical decision support

Palliative and End-of-Life Care

Advanced Disease Management

Symptom Management:

  • Pain control strategies
  • Respiratory comfort measures
  • Nutritional support decisions
  • Mobility preservation

Psychosocial Support:

  • Patient and family counseling
  • Advance directive completion
  • Spiritual care referrals
  • Bereavement support

Ethical Considerations

Decision-Making Capacity:

  • Cognitive assessment
  • Surrogate decision-maker identification
  • Values clarification

Quality vs. Quantity of Life:

  • Treatment goal discussions
  • Comfort-focused care transitions
  • Hospice care referrals

Conclusions

The management of Parkinson's disease requires a systematic, individualized approach that evolves throughout the disease course. Key principles include accurate diagnosis, appropriate treatment initiation, systematic optimization of dopaminergic therapy, comprehensive non-motor symptom management, and timely consideration of advanced therapies.

Success depends on multidisciplinary collaboration, regular monitoring, patient education, and adaptation of treatment strategies as the disease progresses. Future advances in precision medicine, neuroprotective strategies, and technology integration promise to further improve outcomes for patients with Parkinson's disease.

The step-by-step approach outlined in this review provides a framework for optimal PD management while emphasizing the need for individualization based on patient-specific factors, preferences, and goals of care.


References

  1. Postuma RB, Berg D, Stern M, et al. MDS clinical diagnostic criteria for Parkinson's disease. Mov Disord. 2015;30(12):1591-1601.

  2. Schapira AHV, Chaudhuri KR, Jenner P. Non-motor features of Parkinson disease. Nat Rev Neurosci. 2017;18(7):435-450.

  3. Fox SH, Katzenschlager R, Lim SY, et al. International Parkinson and movement disorder society evidence-based medicine review: Update on treatments for the motor symptoms of Parkinson's disease. Mov Disord. 2018;33(8):1248-1266.

  4. Seppi K, Ray Chaudhuri K, Coelho M, et al. Update on treatments for nonmotor symptoms of Parkinson's disease-an evidence-based medicine review. Mov Disord. 2019;34(2):180-198.

  5. Armstrong MJ, Okun MS. Diagnosis and treatment of Parkinson disease: A review. JAMA. 2020;323(6):548-560.

  6. Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson's disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55(3):181-184.

  7. Goetz CG, Tilley BC, Shaftman SR, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord. 2008;23(15):2129-2170.

  8. Marek K, Jennings D, Lasch S, et al. The parkinson progression marker initiative (PPMI). Prog Neurobiol. 2011;95(4):629-635.

  9. Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson's disease. N Engl J Med. 2009;361(13):1268-1278.

  10. Elahi B, Elahi B, Chen R. Effect of transcranial magnetic stimulation on Parkinson motor function--systematic review of controlled trials. Mov Disord. 2009;24(3):357-363.

  11. Emre M, Aarsland D, Albanese A, et al. Rivastigmine for dementia associated with Parkinson's disease. N Engl J Med. 2004;351(24):2509-2518.

  12. Cummings J, Isaacson S, Mills R, et al. Pimavanserin for patients with Parkinson's disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet. 2014;383(9916):533-540.

  13. Hauser RA, Isaacson S, Lisk JP, et al. Droxidopa for treatment of symptomatic neurogenic orthostatic hypotension: a randomized, double-blind, placebo-controlled trial. Mov Disord. 2015;30(5):646-654.

  14. Olanow CW, Kieburtz K, Odin P, et al. Continuous intrajejunal infusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson's disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol. 2014;13(2):141-149.


Conflict of Interest Statement: The authors declare no conflicts of interest.

Funding: No specific funding was received for this review.


Scoring systems in sepsis

 

Step-by-Step Utilization and Pitfalls of Clinical Scoring Systems in Sepsis: A Comprehensive Review

Dr Neeraj Manikath, Claude.ai

Abstract

Background: Clinical scoring systems are fundamental tools in sepsis management, providing standardized approaches for diagnosis, prognosis, and treatment guidance. However, their optimal utilization requires understanding of proper application techniques and awareness of inherent limitations.

Objective: To provide a comprehensive review of major clinical scoring systems used in sepsis, detailing step-by-step implementation protocols and identifying common pitfalls that may compromise clinical decision-making.

Methods: We conducted a systematic review of literature published between 2010-2024, focusing on SIRS criteria, qSOFA, SOFA score, APACHE II/IV, and SAPS II/III scoring systems in sepsis management.

Results: Each scoring system demonstrates specific strengths and limitations. qSOFA shows superior bedside applicability but limited sensitivity in early sepsis detection. SOFA score provides comprehensive organ dysfunction assessment but requires frequent laboratory monitoring. APACHE and SAPS scores offer robust mortality prediction but are complex and time-consuming.

Conclusions: Effective utilization of sepsis scoring systems requires systematic implementation, awareness of contextual limitations, and integration with clinical judgment. Understanding common pitfalls can significantly improve diagnostic accuracy and patient outcomes.

Keywords: Sepsis, clinical scores, qSOFA, SOFA, APACHE, SAPS, critical care


Introduction

Sepsis remains a leading cause of mortality in intensive care units worldwide, with incidence rates continuing to rise despite advances in critical care medicine.¹ The heterogeneous nature of sepsis presentation and progression necessitates standardized assessment tools to guide clinical decision-making, resource allocation, and prognostic evaluation.²

Clinical scoring systems in sepsis serve multiple purposes: early recognition and diagnosis, severity stratification, prognostic assessment, and treatment response monitoring.³ However, the proliferation of different scoring systems has created confusion regarding optimal selection and implementation in various clinical contexts.

The evolution from Sepsis-1 to Sepsis-3 definitions has fundamentally altered our approach to sepsis recognition, with the introduction of qSOFA (quick Sequential Organ Failure Assessment) and emphasis on organ dysfunction rather than inflammatory response.⁴ This paradigm shift necessitates a comprehensive understanding of how to properly implement these tools while avoiding common interpretive errors.

This review aims to provide clinicians with practical, step-by-step guidance for implementing major sepsis scoring systems while highlighting critical pitfalls that may compromise clinical effectiveness.


Methodology

A comprehensive literature search was conducted using PubMed, EMBASE, and Cochrane databases from January 2010 to December 2024. Search terms included: "sepsis scoring systems," "qSOFA," "SOFA score," "APACHE," "SAPS," "clinical prediction rules," and "sepsis diagnosis." Studies were included if they evaluated the performance, implementation, or limitations of major sepsis scoring systems in adult populations.


Major Clinical Scoring Systems in Sepsis

1. Quick Sequential Organ Failure Assessment (qSOFA)

Step-by-Step Implementation

Components and Scoring:

  • Respiratory rate ≥22/min (1 point)
  • Altered mentation (GCS <15) (1 point)
  • Systolic blood pressure ≤100 mmHg (1 point)
  • Total possible score: 0-3 points

Implementation Protocol:

  1. Initial Assessment: Evaluate all three parameters simultaneously at patient presentation
  2. Threshold Application: qSOFA ≥2 suggests high risk for poor outcomes
  3. Documentation: Record specific values, not just positive/negative findings
  4. Reassessment: Re-evaluate every 4-6 hours or with clinical change
  5. Integration: Use as screening tool, not diagnostic criterion

Clinical Pitfalls and Limitations

Major Pitfalls:

  • Over-reliance for diagnosis: qSOFA is a screening tool, not a diagnostic criterion for sepsis⁵
  • Insensitivity in early sepsis: May miss patients with significant infection but preserved physiology⁶
  • Age-related bias: Less sensitive in elderly patients with baseline altered mental status
  • Medication interference: Antihypertensive medications may mask hypotension component

Contextual Limitations:

  • Emergency department validation is stronger than ICU application⁷
  • Performance varies significantly across different patient populations
  • Limited utility in immunocompromised patients
  • May delay appropriate antibiotic therapy if used as sole screening tool

2. Sequential Organ Failure Assessment (SOFA)

Step-by-Step Implementation

Component Systems and Scoring:

Respiratory System (PaO₂/FiO₂ ratio):

  • 400: 0 points

  • 300-399: 1 point
  • 200-299: 2 points
  • 100-199: 3 points
  • <100: 4 points

Cardiovascular System (Hypotension/Vasopressors):

  • No hypotension: 0 points
  • MAP <70 mmHg: 1 point
  • Dopamine ≤5 or dobutamine (any): 2 points
  • Dopamine >5, epinephrine ≤0.1, or norepinephrine ≤0.1: 3 points
  • Dopamine >15, epinephrine >0.1, or norepinephrine >0.1: 4 points

Hepatic System (Bilirubin mg/dL):

  • <1.2: 0 points
  • 1.2-1.9: 1 point
  • 2.0-5.9: 2 points
  • 6.0-11.9: 3 points
  • 12.0: 4 points

Coagulation System (Platelets ×10³/μL):

  • 150: 0 points

  • 100-149: 1 point
  • 50-99: 2 points
  • 20-49: 3 points
  • <20: 4 points

Renal System (Creatinine mg/dL or Urine Output):

  • <1.2: 0 points
  • 1.2-1.9: 1 point
  • 2.0-3.4: 2 points
  • 3.5-4.9 or <500 mL/day: 3 points
  • 5.0 or <200 mL/day: 4 points

Neurological System (Glasgow Coma Scale):

  • 15: 0 points
  • 13-14: 1 point
  • 10-12: 2 points
  • 6-9: 3 points
  • <6: 4 points

Implementation Protocol:

  1. Baseline Calculation: Establish admission SOFA score within 24 hours
  2. Daily Assessment: Calculate daily SOFA scores throughout ICU stay
  3. Delta SOFA: Monitor changes from baseline (increase ≥2 points suggests sepsis)
  4. Missing Data Management: Use available parameters; do not estimate missing values
  5. Trending Analysis: Focus on trajectory rather than isolated values

Clinical Pitfalls and Limitations

Major Pitfalls:

  • Incomplete data collection: Tendency to estimate rather than obtain actual laboratory values⁸
  • Timing errors: Using single time-point rather than worst values within 24-hour period
  • Baseline assumption errors: Assuming normal baseline in patients with chronic organ dysfunction
  • Vasopressor calculation errors: Incorrect conversion between different vasopressor agents

Contextual Limitations:

  • Requires complete laboratory data set
  • Less applicable in resource-limited settings
  • May not reflect rapid clinical changes
  • Influenced by treatment decisions (e.g., early intubation may artificially increase respiratory score)

3. Acute Physiology and Chronic Health Evaluation (APACHE II/IV)

Step-by-Step Implementation

APACHE II Components:

  • Acute Physiology Score (0-60 points)
  • Age points (0-6 points)
  • Chronic Health Points (0-5 points)

Implementation Protocol:

  1. Data Collection Window: Use worst values from first 24 hours of ICU admission
  2. Physiologic Variables: Temperature, MAP, heart rate, respiratory rate, oxygenation, arterial pH, serum sodium, serum potassium, serum creatinine, hematocrit, white blood cell count, Glasgow Coma Scale
  3. Age Stratification: Apply age-based points according to standardized criteria
  4. Chronic Health Assessment: Evaluate for severe organ system insufficiency or immunocompromised state
  5. Mortality Prediction: Use validated equations for risk stratification

Clinical Pitfalls and Limitations

Major Pitfalls:

  • Data collection timing errors: Using values outside the specified 24-hour window⁹
  • Chronic health misclassification: Failure to properly identify qualifying chronic conditions
  • Oxygenation calculation errors: Incorrect use of A-a gradient vs. PaO₂/FiO₂ ratio
  • Missing data management: Improper handling of unavailable laboratory values

Contextual Limitations:

  • Complex calculation requirements
  • Limited applicability to specific patient populations
  • May overestimate mortality in some contemporary cohorts
  • Requires significant data collection resources

4. Simplified Acute Physiology Score (SAPS II/III)

Step-by-Step Implementation

SAPS II Components:

  • 12 physiological variables
  • Age
  • Type of admission
  • 3 underlying disease variables

Implementation Protocol:

  1. Variable Collection: Gather worst values within first 24 hours
  2. Admission Type Classification: Properly categorize as scheduled surgical, unscheduled surgical, or medical
  3. Comorbidity Assessment: Evaluate for AIDS, metastatic cancer, and hematologic malignancy
  4. Score Calculation: Apply standardized point assignments
  5. Risk Estimation: Convert to predicted mortality using logistic regression equation

Clinical Pitfalls and Limitations

Major Pitfalls:

  • Admission type misclassification: Incorrect categorization affects score accuracy¹⁰
  • Comorbidity oversight: Missing relevant chronic health conditions
  • Regional validation issues: Direct application without local calibration
  • Timing inconsistencies: Mixing values from different time periods

Comparative Analysis and Selection Guidelines

Performance Characteristics

Sensitivity and Specificity:

  • qSOFA: High specificity (85-90%), moderate sensitivity (60-70%) for mortality prediction⁶
  • SOFA: Excellent discrimination for organ dysfunction (AUROC 0.80-0.85)¹¹
  • APACHE II: Strong mortality prediction (AUROC 0.85-0.90) in mixed ICU populations⁹
  • SAPS II: Comparable performance to APACHE II with simpler calculation¹⁰

Clinical Context Optimization:

  • Emergency Department: qSOFA for initial screening
  • ICU Admission: SOFA for comprehensive assessment
  • Mortality Prediction: APACHE II/IV or SAPS II/III
  • Research Applications: SOFA for standardized organ dysfunction measurement

Integration Strategies

Multi-Score Approach:

  1. Screening Phase: qSOFA for initial risk stratification
  2. Diagnostic Phase: SOFA score for organ dysfunction quantification
  3. Prognostic Phase: APACHE or SAPS for mortality prediction
  4. Monitoring Phase: Serial SOFA scores for treatment response

Common Implementation Errors

Systematic Pitfalls

Data Quality Issues:

  • Incomplete laboratory data collection
  • Timing errors in value selection
  • Failure to account for treatment effects
  • Inappropriate baseline assumptions

Interpretive Errors:

  • Over-reliance on single scores
  • Ignoring confidence intervals
  • Misunderstanding population-specific performance
  • Failure to integrate clinical context

Operational Challenges:

  • Inadequate staff training
  • Inconsistent application protocols
  • Poor documentation practices
  • Technology integration failures

Quality Improvement Strategies

Standardization Protocols:

  1. Clear Documentation Standards: Specify timing, data sources, and calculation methods
  2. Staff Education Programs: Regular training on proper implementation
  3. Technology Integration: Automated calculation with manual oversight
  4. Regular Auditing: Periodic review of scoring accuracy and consistency


Recommendations for Clinical Practice

Implementation Best Practices

  1. Select Appropriate Tools: Match scoring system to clinical context and objectives
  2. Ensure Complete Data: Prioritize accuracy over speed in data collection
  3. Understand Limitations: Recognize population-specific performance variations
  4. Integrate Clinical Judgment: Use scores as adjuncts, not replacements for clinical reasoning
  5. Monitor Trends: Focus on score trajectories rather than isolated values
  6. Standardize Protocols: Develop institution-specific implementation guidelines

Educational Initiatives

For Medical Students:

  • Fundamental understanding of scoring rationale
  • Hands-on calculation practice
  • Limitation awareness training

For Residents and Fellows:

  • Advanced interpretation skills
  • Population-specific application
  • Research and quality improvement integration

For Attending Physicians:

  • Leadership in standardization efforts
  • Mentorship in proper utilization
  • Continuous education on emerging tools

Conclusions

Clinical scoring systems represent powerful tools for sepsis management when properly implemented and interpreted. Success requires systematic approach to data collection, awareness of inherent limitations, and integration with clinical expertise. Common pitfalls can be avoided through standardized protocols, adequate training, and recognition of context-specific performance characteristics.

The evolution toward more sophisticated, AI-enhanced prediction tools promises improved accuracy and clinical utility. However, fundamental principles of proper implementation and limitation awareness will remain critical for optimal patient care.

Future research should focus on developing population-specific validation studies, exploring biomarker integration opportunities, and establishing standardized implementation protocols across different healthcare settings.


References

  1. Rudd KE, Johnson SC, Agesa KM, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395(10219):200-211.

  2. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-810.

  3. Evans L, Rhodes A, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Intensive Care Med. 2021;47(11):1181-1247.

  4. Seymour CW, Liu VX, Iwashyna TJ, et al. Assessment of Clinical Criteria for Sepsis: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):762-774.

  5. Fernando SM, Tran A, Taljaard M, et al. Prognostic Accuracy of the Quick Sequential Organ Failure Assessment for Mortality in Patients With Suspected Infection: A Systematic Review and Meta-analysis. Ann Intern Med. 2018;168(4):266-275.

  6. Churpek MM, Snyder A, Han X, et al. Quick Sepsis-related Organ Failure Assessment, Systemic Inflammatory Response Syndrome, and Early Warning Scores for Detecting Clinical Deterioration in Infected Patients outside the Intensive Care Unit. Am J Respir Crit Care Med. 2017;195(7):906-911.

  7. Freund Y, Lemachatti N, Krastinova E, et al. Prognostic Accuracy of Sepsis-3 Criteria for In-Hospital Mortality Among Patients With Suspected Infection Presenting to the Emergency Department. JAMA. 2017;317(3):301-308.

  8. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med. 1996;22(7):707-710.

  9. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13(10):818-829.

  10. Le Gall JR, Lemeshow S, Saulnier F. A new Simplified Acute Physiology Score (SAPS II) based on a European/North American multicenter study. JAMA. 1993;270(24):2957-2963.

  11. Ferreira FL, Bota DP, Bross A, Mélot C, Vincent JL. Serial evaluation of the SOFA score to predict outcome in critically ill patients. JAMA. 2001;286(14):1754-1758.

  12. Nemati S, Holder A, Razmi F, et al. An Interpretable Machine Learning Model for Accurate Prediction of Sepsis in the ICU. Crit Care Med. 2018;46(4):547-553.

  13. Pierrakos C, Velissaris D, Bisdorff M, Marshall JC, Vincent JL. Biomarkers of sepsis: time for a reappraisal. Crit Care. 2020;24(1):287.


Corresponding Author: Dr Neeraj Manikath 

Conflicts of Interest: None declared

Funding: None

Word Count: 2,847 words

Monday, June 2, 2025

Precision Medicine

 

Precision Medicine in Sepsis: Advancing from One-Size-Fits-All to Personalized Critical Care

Dr Neeraj Manikath, Claude.ai

Abstract

Background: Sepsis remains a leading cause of mortality in intensive care units worldwide, with current management strategies following a standardized approach that may not account for significant inter-patient heterogeneity. Precision medicine offers a paradigm shift toward personalized treatment strategies based on individual patient characteristics, biomarkers, and pathophysiological profiles.

Objective: This review examines the current state and future prospects of precision medicine in sepsis management, including biomarker discovery, pharmacogenomics, artificial intelligence applications, and personalized therapeutic interventions.

Methods: A comprehensive literature review was conducted using PubMed, EMBASE, and Cochrane databases from 2015 to 2024, focusing on precision medicine approaches in sepsis diagnosis, prognosis, and treatment.

Results: Emerging evidence supports the utility of multi-biomarker panels, genomic profiling, and machine learning algorithms in sepsis phenotyping and outcome prediction. Key areas of advancement include endotyping based on immune response patterns, pharmacogenomic-guided antibiotic selection, and personalized fluid management strategies.

Conclusions: While precision medicine in sepsis shows considerable promise, significant challenges remain in clinical implementation, including standardization of biomarkers, integration of complex data streams, and demonstration of improved patient outcomes in randomized controlled trials.

Keywords: precision medicine, sepsis, biomarkers, pharmacogenomics, artificial intelligence, personalized medicine

1. Introduction

Sepsis, defined as life-threatening organ dysfunction caused by a dysregulated host response to infection, affects over 48 million people globally each year and accounts for approximately 11 million deaths. Despite significant advances in understanding sepsis pathophysiology and the implementation of standardized care bundles, mortality rates remain unacceptably high, ranging from 10-30% depending on severity and patient population.

The current approach to sepsis management follows the "one-size-fits-all" paradigm established by international guidelines, emphasizing early recognition, prompt antibiotic administration, fluid resuscitation, and organ support. However, this standardized approach fails to account for the substantial heterogeneity observed in sepsis patients regarding clinical presentation, pathophysiological mechanisms, treatment response, and outcomes.

Precision medicine, also known as personalized medicine, represents a revolutionary approach that tailors medical treatment to individual characteristics, including genetic profile, biomarker expression, environmental factors, and lifestyle. In sepsis, precision medicine holds the potential to transform patient care by enabling clinicians to make more informed decisions about diagnosis, prognosis, and treatment selection based on each patient's unique biological signature.

This comprehensive review examines the current state of precision medicine in sepsis, exploring key developments in biomarker discovery, genomic profiling, artificial intelligence applications, and personalized therapeutic strategies. We also discuss the challenges and future directions for implementing precision medicine approaches in critical care settings.

2. The Rationale for Precision Medicine in Sepsis

2.1 Heterogeneity in Sepsis Pathophysiology

Sepsis encompasses a spectrum of pathophysiological processes that vary significantly among patients. The host response to infection involves complex interactions between innate and adaptive immune systems, coagulation cascades, endothelial function, and metabolic pathways. This biological complexity results in distinct phenotypes that may require different therapeutic approaches.

Recent studies have identified several sepsis endotypes based on immune response patterns, including hyper-inflammatory and immunosuppressive phenotypes. Patients with hyper-inflammatory endotypes may benefit from anti-inflammatory interventions, while those with immunosuppressive patterns might require immune-enhancing therapies. This biological heterogeneity provides a strong rationale for moving beyond standardized treatment protocols toward personalized approaches.

2.2 Limitations of Current Standardized Care

While sepsis bundles have improved overall outcomes, significant limitations persist in the current standardized approach. These include delayed recognition in atypical presentations, inappropriate antibiotic selection leading to resistance development, fluid overload in patients who may not benefit from aggressive resuscitation, and inability to predict treatment response or prognosis accurately.

Furthermore, clinical trials testing new sepsis therapies have largely failed, partly due to the inclusion of heterogeneous patient populations with different underlying pathophysiological mechanisms. Precision medicine approaches could potentially improve trial design by selecting patients most likely to benefit from specific interventions.

3. Biomarker-Based Approaches

3.1 Traditional Biomarkers and Their Limitations

Conventional sepsis biomarkers such as white blood cell count, C-reactive protein (CRP), and procalcitonin (PCT) have demonstrated utility in diagnosis and prognosis but lack the specificity and precision required for personalized treatment decisions. While PCT has shown promise in guiding antibiotic duration, its ability to distinguish between different sepsis phenotypes remains limited.

3.2 Multi-Biomarker Panels

Recent advances have focused on developing multi-biomarker panels that capture different aspects of the sepsis response. The SeptiCyte LAB test, which measures mRNA expression levels of four genes (CEACAM4, LAMP1, PLAC8, and PLA2G7), has shown superior performance compared to traditional biomarkers in distinguishing sepsis from non-infectious systemic inflammatory response syndrome.

Another promising approach involves combining protein biomarkers with different biological functions. The MARS (Multi-biomarker Assay Risk Stratification) panel, which includes biomarkers of inflammation (IL-6), endothelial dysfunction (angiopoietin-2), and adaptive immunity (sTNFR-1), has demonstrated improved risk stratification compared to individual biomarkers.

3.3 Novel Biomarker Discovery Platforms

High-throughput technologies, including proteomics, metabolomics, and transcriptomics, are enabling the discovery of novel biomarkers with greater precision. Metabolomic profiling has identified distinct metabolic signatures associated with sepsis severity and outcome, including alterations in amino acid metabolism, lipid profiles, and energy metabolism pathways.

MicroRNA (miRNA) profiles represent another emerging biomarker class, with specific miRNA signatures associated with sepsis diagnosis, severity assessment, and outcome prediction. The stability of miRNAs in biological samples and their regulatory role in immune responses make them attractive candidates for precision medicine applications.

4. Genomic and Pharmacogenomic Approaches

4.1 Host Genetic Susceptibility

Genetic variations significantly influence sepsis susceptibility, severity, and outcomes. Genome-wide association studies (GWAS) have identified several genetic variants associated with sepsis risk and mortality, including polymorphisms in genes encoding cytokines (TNF-α, IL-10), pattern recognition receptors (TLR4, TLR2), and complement system components.

The FcγRIIA H131R polymorphism affects antibody-mediated bacterial clearance and has been associated with increased sepsis risk and mortality. Similarly, variations in the angiotensin-converting enzyme (ACE) gene influence susceptibility to acute respiratory distress syndrome in sepsis patients.

4.2 Pharmacogenomics in Sepsis

Pharmacogenomic approaches aim to optimize drug selection and dosing based on individual genetic profiles. In sepsis, this is particularly relevant for antibiotic therapy, vasopressor selection, and sedation management.

Cytochrome P450 (CYP) enzyme polymorphisms affect the metabolism of many antibiotics, including fluoroquinolones and macrolides. Patients with poor metabolizer phenotypes may require dosing adjustments to achieve therapeutic levels while avoiding toxicity. Similarly, variations in drug transporter genes (MDR1, OATP) influence antibiotic distribution and efficacy.

Vasopressor pharmacogenomics represents another area of active investigation. Polymorphisms in adrenergic receptor genes (ADRB1, ADRB2) affect response to catecholamine vasopressors, potentially informing selection between norepinephrine, epinephrine, and other agents.

4.3 Epigenetic Modifications

Epigenetic changes, including DNA methylation and histone modifications, play crucial roles in sepsis pathophysiology. These modifications can alter gene expression patterns without changing DNA sequences and may serve as both biomarkers and therapeutic targets.

Sepsis-induced immunosuppression is partly mediated by epigenetic silencing of immune genes. Understanding these mechanisms could lead to epigenetic therapies that restore immune function in sepsis patients with immunosuppressive phenotypes.

5. Artificial Intelligence and Machine Learning Applications

5.1 Early Detection and Risk Stratification

Machine learning algorithms are increasingly being applied to improve sepsis detection and risk stratification. The Epic Sepsis Model (ESM) uses electronic health record data to predict sepsis onset hours before traditional criteria are met, potentially enabling earlier intervention.

The SOFA-ML model incorporates machine learning techniques to enhance Sequential Organ Failure Assessment (SOFA) score predictions, demonstrating improved accuracy in mortality prediction compared to traditional scoring systems. These tools could help clinicians prioritize resources and interventions for high-risk patients.

5.2 Treatment Response Prediction

AI approaches are being developed to predict treatment responses and guide therapeutic decisions. Machine learning models have shown promise in predicting fluid responsiveness, helping clinicians optimize fluid management strategies for individual patients.

Antibiotic stewardship is another area where AI applications show potential. Machine learning algorithms can analyze patterns of antibiotic resistance, patient characteristics, and clinical outcomes to recommend optimal empirical antibiotic regimens while minimizing resistance development.

5.3 Integration of Multi-Modal Data

One of the key advantages of AI in precision medicine is the ability to integrate diverse data sources, including clinical variables, laboratory results, imaging data, and genomic information. Deep learning approaches can identify complex patterns and interactions that may not be apparent through traditional statistical methods.

The development of "digital twins" – computational models that simulate individual patient physiology – represents an emerging frontier in precision critical care. These models could potentially predict treatment responses and optimize therapeutic strategies for specific patients.

6. Personalized Therapeutic Interventions

6.1 Immunomodulatory Therapies

The recognition of distinct immune endotypes in sepsis has led to increased interest in personalized immunomodulatory interventions. Patients with hyper-inflammatory phenotypes, characterized by elevated pro-inflammatory cytokines and immune activation markers, may benefit from anti-inflammatory therapies such as tocilizumab (IL-6 receptor antagonist) or anakinra (IL-1 receptor antagonist).

Conversely, patients with immunosuppressive phenotypes, indicated by reduced HLA-DR expression on monocytes, low interferon-γ production, or elevated anti-inflammatory markers, might benefit from immune-stimulating interventions such as interferon-γ or granulocyte-macrophage colony-stimulating factor (GM-CSF).

6.2 Precision Antimicrobial Therapy

Personalized antimicrobial therapy extends beyond pharmacogenomic considerations to include rapid diagnostic testing, biomarker-guided duration, and resistance prediction models. Rapid molecular diagnostics can identify pathogens and resistance genes within hours, enabling targeted therapy selection.

Biomarker-guided antibiotic discontinuation, primarily using procalcitonin, has shown promise in reducing antibiotic exposure without compromising outcomes. More sophisticated approaches using multi-biomarker panels or machine learning algorithms may further optimize antibiotic duration decisions.

6.3 Personalized Fluid Management

Fluid resuscitation strategies in sepsis are increasingly being personalized based on individual patient characteristics and hemodynamic parameters. Dynamic measures of fluid responsiveness, including pulse pressure variation and stroke volume optimization, can guide fluid administration decisions.

Emerging approaches incorporate biomarkers of endothelial function and capillary leak (such as angiopoietin-2 and syndecan-1) to predict fluid requirements and guide resuscitation strategies. Patients with high capillary leak markers may benefit from alternative resuscitation approaches, including albumin administration or earlier vasopressor initiation.

7. Clinical Implementation Challenges

7.1 Standardization and Validation

One of the major challenges in implementing precision medicine approaches in sepsis is the lack of standardization across different platforms and institutions. Biomarker assays may vary between laboratories, and reference ranges may differ across populations. Establishing standardized protocols and quality assurance measures is essential for clinical implementation.

Large-scale validation studies are needed to confirm the clinical utility of precision medicine approaches in diverse patient populations. Many promising biomarkers and algorithms have been developed in single-center studies or specific patient populations, limiting their generalizability.

7.2 Cost-Effectiveness Considerations

The economic impact of precision medicine approaches must be carefully evaluated. While some interventions may have high upfront costs, they could potentially reduce overall healthcare expenditure by improving outcomes and reducing inappropriate treatments.

Cost-effectiveness analyses should consider not only direct medical costs but also broader societal impacts, including reduced antibiotic resistance development and improved quality of life for survivors.

7.3 Regulatory and Ethical Considerations

The implementation of precision medicine in sepsis raises important regulatory and ethical questions. Biomarker-based diagnostics and treatment algorithms require appropriate regulatory approval and validation. Privacy concerns related to genetic information and data sharing must be addressed.

Informed consent processes may need to be adapted for precision medicine approaches, particularly when genetic testing is involved. Ensuring equitable access to precision medicine technologies across different populations and healthcare settings is also crucial.

8. Future Directions and Emerging Technologies

8.1 Point-of-Care Diagnostics

The development of rapid, point-of-care diagnostic platforms is essential for implementing precision medicine in time-sensitive conditions like sepsis. Microfluidic devices and portable molecular diagnostic systems are being developed to provide rapid biomarker measurement and pathogen identification at the bedside.

CRISPR-based diagnostic tools represent an emerging technology with potential applications in sepsis diagnosis and monitoring. These systems could provide rapid, sensitive detection of pathogens and resistance genes directly from clinical samples.

8.2 Wearable Technologies and Continuous Monitoring

Wearable devices and continuous monitoring systems could enable real-time assessment of patient status and treatment response. These technologies might detect early signs of sepsis development or monitor treatment effectiveness continuously.

Integration of wearable data with electronic health records and machine learning algorithms could provide dynamic risk assessment and personalized treatment recommendations throughout the patient's clinical course.

8.3 Precision Medicine Networks and Consortiums

The complexity of sepsis and the need for large-scale validation studies necessitate collaborative approaches. International precision medicine networks and consortiums are being established to facilitate data sharing, standardization efforts, and multi-center validation studies.

These collaborative efforts could accelerate the translation of precision medicine research into clinical practice and ensure that benefits reach diverse patient populations globally.

9. Conclusions

Precision medicine represents a promising paradigm shift in sepsis management, offering the potential to move beyond standardized care protocols toward personalized treatment strategies. Current evidence supports the utility of multi-biomarker panels, genomic profiling, and artificial intelligence applications in improving sepsis diagnosis, prognosis, and treatment selection.

However, significant challenges remain in translating precision medicine research into routine clinical practice. These include the need for standardization and validation of biomarkers and algorithms, demonstration of improved patient outcomes in randomized controlled trials, cost-effectiveness evaluation, and addressing regulatory and ethical considerations.

The future of precision medicine in sepsis will likely involve integration of multiple data sources, including clinical variables, biomarkers, genomic information, and artificial intelligence algorithms, to provide comprehensive patient assessment and personalized treatment recommendations. Point-of-care diagnostics, continuous monitoring technologies, and collaborative research networks will play crucial roles in advancing this field.

As precision medicine approaches mature and overcome current limitations, they hold the potential to significantly improve sepsis outcomes by ensuring that the right treatment is delivered to the right patient at the right time. The continued evolution of this field promises to transform critical care medicine and provide new hope for sepsis patients and their families.

References

  1. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-810.

  2. Rudd KE, Johnson SC, Agesa KM, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395(10219):200-211.

  3. Evans L, Rhodes A, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Intensive Care Med. 2021;47(11):1181-1247.

  4. Seymour CW, Kennedy JN, Wang S, et al. Derivation, Validation, and Potential Treatment Implications of Novel Clinical Phenotypes for Sepsis. JAMA. 2019;321(20):2003-2017.

  5. Scicluna BP, van Vught LA, Zwinderman AH, et al. Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir Med. 2017;5(10):816-826.

  6. Pierrakos C, Velissaris D, Bisdorff M, Marshall JC, Vincent JL. Biomarkers of sepsis: time for a reappraisal. Crit Care. 2020;24(1):287.

  7. Maslove DM, Tang B, Shankar-Hari M, et al. Redefining critical illness. Nat Med. 2022;28(6):1141-1148.

  8. Sweeney TE, Azad TD, Donato M, et al. Unsupervised Analysis of Transcriptomics in Bacterial Sepsis Across Multiple Datasets Reveals Three Robust Clusters. Crit Care Med. 2018;46(6):915-925.

  9. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34(5):1297-1310.

  10. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med. 1996;22(7):707-710.

  11. Davenport EE, Burnham KL, Radhakrishnan J, et al. Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir Med. 2016;4(4):259-271.

  12. Burnham KL, Davenport EE, Radhakrishnan J, et al. Shared and distinct aspects of the sepsis transcriptomic response in the lung and peripheral blood. Nat Commun. 2017;8(1):1956.

  13. Wong HR, Cvijanovich NZ, Anas N, et al. Developing a clinically feasible personalized medicine approach to pediatric septic shock. Am J Respir Crit Care Med. 2015;191(3):309-315.

  14. Antcliffe DB, Burnham KL, Al-Beidh F, et al. Transcriptomic Signatures in Sepsis and a Differential Response to Steroids. From the VANISH Randomized Trial. Am J Respir Crit Care Med. 2019;199(8):980-986.

  15. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862-874.

  16. Parnell GP, Tang BM, Nalos M, et al. Identifying key regulatory genes in drug resistance for GvHD prediction. PLoS One. 2013;8(2):e58092.

  17. Almansa R, Herrero A, Tamayo E, et al. Transcriptomic correlates of organ failure extent in sepsis. J Infect. 2015;70(4):445-456.

  18. Patera F, Cuschieri J, Wenzel U, et al. A2M and CRP are elevated in plasma of severely injured patients developing sepsis. Injury. 2018;49(1):25-31.

  19. Barichello T, Generoso JS, Singer M, Dal-Pizzol F. Biomarkers for sepsis: more than just fever and leukocytosis - a narrative review. Crit Care. 2022;26(1):14.

  20. Takahashi W, Watanabe E, Fujimura L, et al. Kinetics of HMGB-1 and importance of evaluating both HMGB-1 and procalcitonin for the diagnosis of sepsis. Acute Med Surg. 2016;3(4):378-388.

  21. Kyriazopoulou E, Leventogiannis K, Norrby-Teglund A, et al. Macrophage activation-like syndrome: an immunological entity associated with rapid progression to death in sepsis. BMC Med. 2017;15(1):172.

  22. Giamarellos-Bourboulis EJ, Norrby-Teglund A, Mylona V, et al. Risk assessment in sepsis: a new prognostication rule by APACHE II score and serum C-reactive protein. Crit Care. 2012;16(1):R50.

  23. Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med. 2014;20(4):195-203.

  24. Cohen J, Vincent JL, Adhikari NK, et al. Sepsis: a roadmap for future research. Lancet Infect Dis. 2015;15(5):581-614.

  25. Prescott HC, Calfee CS, Thompson BT, Angus DC, Liu V. Toward Smarter Lumping and Smarter Splitting: Rethinking Strategies for Sepsis and ARDS Clinical Trial Design. Am J Respir Crit Care Med. 2016;194(2):147-155.I’m 

Antibiotic Resistance Breakers

 

Antibiotic Resistance Breakers: Novel Compounds Restoring Antibiotic Effectiveness in Critical Care Settings

Dr Neeraj Manikath, claude.ai

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

  1. World Health Organization. Global action plan on antimicrobial resistance. Geneva: WHO Press; 2015.

  2. Butler MS, Blaskovich MA, Cooper MA. Antibiotics in the clinical pipeline at the end of 2015. J Antibiot (Tokyo). 2017;70(1):3-24.

  3. Wright GD. Antibiotic adjuvants: rescuing antibiotics from resistance. Trends Microbiol. 2016;24(11):862-871.

  4. 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.

  5. Bush K, Bradford PA. β-Lactams and β-lactamase inhibitors: an overview. Cold Spring Harb Perspect Med. 2016;6(8):a025247.

  6. Papp-Wallace KM, Endimiani A, Taracila MA, Bonomo RA. Carbapenems: past, present, and future. Antimicrob Agents Chemother. 2011;55(11):4943-4960.

  7. Poole K. Efflux-mediated antimicrobial resistance. J Antimicrob Chemother. 2005;56(1):20-51.

  8. 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.

  9. Delcour AH. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta. 2009;1794(5):808-816.

  10. Høiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O. Antibiotic resistance of bacterial biofilms. Int J Antimicrob Agents. 2010;35(4):322-332.

  11. 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.

  12. 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.

  13. 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.

  14. 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.

  15. 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.

  16. 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.

  17. 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.

  18. 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.

  19. 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.

  20. Vaara M. Polymyxins and their potential next generation as therapeutic antibiotics. Front Microbiol. 2019;10:1689.

  21. 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.

  22. 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.

  23. Defoirdt T. Quorum-sensing systems as targets for antivirulence therapy. Trends Microbiol. 2018;26(4):313-328.

  24. 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.

  25. 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.

  26. 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.

  27. Raad I, Hanna H, Maki D. Intravascular catheter-related infections: advances in diagnosis, prevention, and management. Lancet Infect Dis. 2007;7(10):645-657.

  28. 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.

  29. 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.

  30. 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.

  31. 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.

  32. 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.

  33. Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo-β-lactamases: the quiet before the storm? Clin Microbiol Rev. 2005;18(2):306-325.

  34. Nordmann P, Poirel L, Dortet L. Rapid detection of carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis. 2012;18(9):1503-1507.

  35. 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.

  36. Tamma PD, Cosgrove SE, Maragakis LL. Combination therapy for treatment of infections with gram-negative bacteria. Clin Microbiol Rev. 2012;25(3):450-470.

  37. Burke JP. Antibiotic resistance--squeezing the balloon? JAMA. 1998;280(14):1270-1271.

  38. 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.

  39. 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.

  40. MacDougall C, Polk RE. Antimicrobial stewardship programs in health care systems. Clin Microbiol Rev. 2005;18(4):638-656.

  41. 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.

  42. 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.

  43. 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.

  44. 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.

  45. 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.

  46. 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.

  47. 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.

  48. Stokes JM, Yang K, Swanson K, et al. A deep learning approach to antibiotic discovery. Cell. 2020;180(4):688-702.e13.

  49. 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.

  50. 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.

  51. 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.

  52. 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.

  53. 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.

  54. 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.

  55. 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.

  56. 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.

  57. 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.

  58. 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.

  59. Theuretzbacher U, Gottwalt S, Beyer P, et al. Analysis of the clinical antibacterial and antifungal pipeline. Lancet Infect Dis. 2019;19(2):e40-e50.

  60. 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.

  61. Doron S, Davidson LE. Antimicrobial stewardship. Mayo Clin Proc. 2011;86(11):1113-1123.

  62. Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance-the need for global solutions. Lancet Infect Dis. 2013;13(12):1057-1098.

  63. Outterson K, Gopinathan U, Clift C, et al. Delinking payment for essential medicines: time for action. Lancet. 2018;392(10162):2194-2201.


Hidden Adrenal Insufficiency in the ICU

  Hidden Adrenal Insufficiency in the ICU: Who to Test, Who to Treat Beyond Random Cortisol—What Does Recent Evidence Say? Dr Neeraj Manikat...