Tuesday, September 16, 2025

Emerging Antivirals in ICU Practice

 

Emerging Antivirals in ICU Practice: A Critical Care Perspective on Remdesivir, Nirmatrelvir/Ritonavir, and Newer Broad-Spectrum Agents

Dr Neeraj Manikath , claude.ai

Abstract

The landscape of antiviral therapeutics in critical care has evolved dramatically following the COVID-19 pandemic, with significant implications for intensive care unit (ICU) practice. This review examines the current evidence, clinical applications, and practical considerations for emerging antivirals including remdesivir, nirmatrelvir/ritonavir (Paxlovid), and newer broad-spectrum agents. We analyze their mechanisms of action, pharmacokinetics, efficacy data, safety profiles, and integration into critical care protocols. Special emphasis is placed on drug interactions, dosing considerations in organ dysfunction, and emerging resistance patterns. Clinical pearls and practical insights are provided to guide intensivists in optimizing antiviral therapy for critically ill patients.

Keywords: antivirals, critical care, remdesivir, nirmatrelvir/ritonavir, COVID-19, intensive care unit


Introduction

The emergence of SARS-CoV-2 catalyzed unprecedented development and deployment of antiviral therapeutics in critical care settings. Beyond COVID-19, the intensivist must now navigate an expanding armamentarium of antiviral agents with diverse mechanisms, indications, and limitations. This evolution represents a paradigm shift from the historically limited antiviral options available for critically ill patients to a more nuanced, pathogen-specific approach.

The critical care environment presents unique challenges for antiviral therapy implementation, including altered pharmacokinetics due to organ dysfunction, complex drug interactions with standard ICU medications, and the need for rapid therapeutic decisions in the face of clinical deterioration. This review provides a comprehensive analysis of current and emerging antivirals relevant to ICU practice, with emphasis on evidence-based application and practical considerations.


Remdesivir: The Prototype Direct-Acting Antiviral

Mechanism of Action and Pharmacology

Remdesivir (GS-5734) is a nucleotide analog prodrug that targets viral RNA-dependent RNA polymerases (RdRp). Following intracellular conversion to its active triphosphate metabolite (GS-443902), it acts as an RNA chain terminator, effectively halting viral replication across multiple RNA viruses including coronaviruses, filoviruses, and paramyxoviruses.

The pharmacokinetic profile of remdesivir is particularly relevant to critical care practice. The drug exhibits rapid plasma clearance (t₁/₂ = 1 hour) but demonstrates prolonged intracellular retention of active metabolites (t₁/₂ = 14-35 hours in peripheral blood mononuclear cells). This dichotomy between plasma and intracellular kinetics influences dosing strategies and clinical efficacy.

Clinical Evidence and Efficacy

The ACTT-1 trial established remdesivir's role in COVID-19 management, demonstrating a 31% reduction in time to recovery (median 10 vs. 15 days, p<0.001) in hospitalized patients. Subsequent analyses revealed greatest benefit in patients requiring supplemental oxygen without mechanical ventilation, with diminished efficacy in those requiring high-flow oxygen or mechanical ventilation.

Pearl: The efficacy of remdesivir appears inversely related to disease severity, suggesting optimal timing is during the viremic phase before extensive inflammatory cascades develop.

Critical care-specific data from the ACTT-1 subgroup analysis showed:

  • Mechanically ventilated patients: Rate ratio for recovery 0.95 (95% CI: 0.64-1.42)
  • Patients on high-flow oxygen: Rate ratio 1.09 (95% CI: 0.76-1.57)
  • Mortality reduction was not statistically significant in critically ill cohorts

Dosing and Administration in Critical Care

Standard Dosing Protocol:

  • Loading dose: 200 mg IV on day 1
  • Maintenance: 100 mg IV daily for 4-9 days (total course 5-10 days)
  • Infusion rate: ≤4 mg/min to minimize infusion reactions

Critical Care Considerations:

Renal Impairment: Remdesivir is contraindicated in eGFR <30 mL/min/1.73m² due to accumulation of the sulfobutylether-β-cyclodextrin (SBECD) excipient, which may cause nephrotoxicity.

Hepatic Dysfunction: Dose reduction is not routinely recommended, but careful monitoring of hepatic transaminases is essential given reports of drug-induced liver injury.

Continuous Renal Replacement Therapy (CRRT): Limited data suggest remdesivir may be administered during CRRT, though active metabolite clearance remains uncertain.

Safety Profile and Monitoring

Common Adverse Effects:

  • Hepatotransaminase elevation (8-10% of patients)
  • Infusion-related reactions (hypotension, nausea, diaphoresis)
  • Bradycardia (particularly with rapid infusion)
  • Acute kidney injury (1-3% incidence)

Monitoring Parameters:

  • Daily liver function tests for first 3 days, then every 48 hours
  • Serum creatinine and eGFR daily
  • Prothrombin time (remdesivir may prolong PT/INR)

Oyster: Apparent clinical improvement following remdesivir initiation may mask underlying hepatotoxicity. Always check LFTs before attributing clinical deterioration to disease progression alone.


Nirmatrelvir/Ritonavir (Paxlovid): Protease Inhibition in Critical Care

Mechanism and Rationale

Nirmatrelvir is a potent, selective inhibitor of the SARS-CoV-2 main protease (3CLpro), essential for viral polyprotein processing and replication. Co-formulated ritonavir serves as a pharmacokinetic enhancer, inhibiting CYP3A4-mediated metabolism of nirmatrelvir and extending its half-life from 6.2 to 13.1 hours.

This combination represents a shift toward oral antiviral therapy, though its application in critical care is complicated by drug interaction potential and contraindications in severe illness.

Clinical Evidence

The EPIC-HR trial demonstrated remarkable efficacy in high-risk, non-hospitalized patients with 89% reduction in severe COVID-19 outcomes when initiated within 3 days of symptom onset. However, critical care applications are limited by study exclusion criteria and subsequent real-world evidence.

Key Efficacy Data:

  • Primary endpoint reduction: 89% (95% CI: 75-96%)
  • Hospitalization reduction: 6.3% vs. 0.77% (absolute risk reduction 5.8%)
  • Mortality reduction: 12 deaths (placebo) vs. 0 deaths (nirmatrelvir/ritonavir)

Critical Care Limitations:

  • Limited data in mechanically ventilated patients
  • Contraindicated with many ICU medications
  • Oral administration challenges in critically ill patients

Drug Interactions: The Critical Care Challenge

Ritonavir is a potent inhibitor of CYP3A4, CYP2D6, and P-glycoprotein, creating extensive interaction potential with standard ICU medications.

Major Contraindicated Drugs in ICU Settings:

  • Sedatives: Midazolam (IV formulations), triazolam
  • Analgesics: Pethidine, tramadol, propoxyphene
  • Cardiovascular: Amiodarone, dronedarone, flecainide, propafenone
  • Anticoagulants: Direct interaction with warfarin metabolism
  • Vasopressors: Potential interactions with ergot alkaloids

Drugs Requiring Dose Adjustment:

  • Dexamethasone: Increase monitoring for hyperglycemia
  • Tacrolimus: Reduce dose by 75% and monitor levels closely
  • Atorvastatin: Temporarily discontinue or reduce dose
  • Calcium channel blockers: Monitor for hypotension

Hack: Create an ICU-specific drug interaction checklist for nirmatrelvir/ritonavir. Many interactions can be managed with temporary dose adjustments rather than absolute contraindications.

Practical Implementation in Critical Care

Patient Selection Criteria:

  • Able to tolerate oral medications or have functioning enteral access
  • Less than 5 days from symptom onset
  • Absence of major drug interactions
  • eGFR ≥30 mL/min/1.73m² (standard dose)

Dosing Modifications:

  • Standard: Nirmatrelvir 300 mg + ritonavir 100 mg twice daily × 5 days
  • Moderate renal impairment (eGFR 30-60): Nirmatrelvir 150 mg + ritonavir 100 mg twice daily
  • Severe renal impairment (eGFR <30): Contraindicated

Administration Considerations:

  • Must be given with food to enhance absorption
  • Complete 5-day course even if symptoms resolve
  • Cannot be crushed or divided (film-coated tablets)

Newer Broad-Spectrum Antivirals: Expanding the Arsenal

Molnupiravir: The Mutagenic Approach

Molnupiravir (β-D-N4-hydroxycytidine) represents a novel mechanism of antiviral action through lethal mutagenesis. The active metabolite, β-D-N4-hydroxycytidine triphosphate, is incorporated into viral RNA, causing error catastrophe and viral extinction.

Clinical Evidence: The MOVe-OUT trial showed modest benefit with 30% reduction in hospitalization or death among high-risk, non-hospitalized COVID-19 patients. However, the benefit was less pronounced than nirmatrelvir/ritonavir, and mutagenic concerns limit its use in certain populations.

Critical Care Applications:

  • Limited by modest efficacy data
  • Oral administration only
  • Contraindicated in pregnancy due to mutagenic potential
  • Fewer drug interactions compared to ritonavir-boosted regimens

Bebtelovimab: Monoclonal Antibody Therapy

While technically not a small-molecule antiviral, bebtelovimab represents an important therapeutic option for immunocompromised critically ill patients who may not mount adequate immune responses to standard antivirals.

Advantages in Critical Care:

  • Single IV dose administration
  • Maintains activity against most Omicron variants
  • Particularly valuable in immunocompromised patients
  • Minimal drug interactions

Limitations:

  • Variant susceptibility changes
  • Limited supply and high cost
  • Requires IV administration capability

Emerging Broad-Spectrum Agents

GS-5245 (Obeldesivir): A next-generation nucleotide analog with improved bioavailability and broader spectrum activity. Early phase studies suggest potential advantages over remdesivir in terms of oral bioavailability and reduced nephrotoxicity risk.

VV116 (JT001): An oral nucleoside analog showing non-inferiority to nirmatrelvir/ritonavir in early studies, with potentially fewer drug interactions due to lack of ritonavir boosting.


Integration into ICU Protocols

Treatment Algorithm Development

Early Recognition and Rapid Deployment: Successful antiviral therapy in critical care requires rapid pathogen identification and treatment initiation. Point-of-care molecular diagnostics and standardized treatment algorithms are essential.

Proposed ICU Antiviral Decision Tree:

  1. Rapid Diagnostic Testing (within 4 hours of ICU admission)
  2. Symptom Duration Assessment (<5 days optimal for most agents)
  3. Drug Interaction Screening (automated systems recommended)
  4. Renal/Hepatic Function Assessment
  5. Route of Administration Feasibility (IV vs. oral)

Monitoring and Optimization

Therapeutic Drug Monitoring: While routine TDM is not established for most antivirals, consider monitoring in patients with:

  • Significant organ dysfunction
  • Suspected drug interactions
  • Clinical treatment failure
  • Prolonged treatment courses

Clinical Response Assessment:

  • Viral load monitoring (when available)
  • Clinical symptom scoring systems
  • Inflammatory marker trends (CRP, procalcitonin, ferritin)
  • Oxygenation indices and ventilator weaning parameters

Combination Therapy Considerations

Pearl: Combination antiviral therapy should generally be avoided outside of clinical trials due to:

  • Lack of established efficacy data
  • Potential for additive toxicities
  • Increased drug interaction complexity
  • Cost considerations

Exception: Immunocompromised patients may benefit from combination approaches, but this should be individualized and ideally coordinated with infectious disease specialists.


Special Populations in Critical Care

Immunocompromised Patients

Immunocompromised critically ill patients present unique challenges requiring modified antiviral approaches:

Extended Treatment Duration:

  • Standard 5-day courses may be insufficient
  • Consider 10-14 day courses based on viral clearance
  • Weekly viral load monitoring when available

Combination Strategies:

  • Antiviral + monoclonal antibody therapy
  • Sequential antiviral therapy if resistance develops
  • Convalescent plasma as adjunctive therapy

Pregnancy and Lactation

Remdesivir: Preferred agent based on extensive safety data Nirmatrelvir/Ritonavir: Limited data; use only if benefits outweigh risks Molnupiravir: Contraindicated due to mutagenic potential

Hack: For pregnant patients in ICU, establish multidisciplinary team including maternal-fetal medicine, infectious disease, and critical care specialists before treatment initiation.

Pediatric Critical Care

Pediatric dosing and safety data remain limited for newer antivirals:

Remdesivir Pediatric Dosing:

  • Weight-based dosing for patients <40 kg
  • Loading dose: 5 mg/kg IV
  • Maintenance: 2.5 mg/kg IV daily

Resistance and Future Considerations

Resistance Mechanisms

Understanding resistance patterns is crucial for optimizing therapy and anticipating treatment failures:

Remdesivir Resistance:

  • RdRp mutations (F480L, V557L, A550V)
  • Generally associated with reduced fitness
  • Cross-resistance with other nucleoside analogs possible

Nirmatrelvir Resistance:

  • 3CLpro mutations (E166V, L50F, K90R)
  • May emerge rapidly in immunocompromised patients
  • Potential for cross-resistance with other protease inhibitors

Clinical Implications:

  • Sequential viral load monitoring in high-risk patients
  • Consider resistance testing if treatment failure suspected
  • Alternative therapy planning for resistant isolates

Future Therapeutic Targets

Host-Directed Therapies:

  • Targeting cellular pathways essential for viral replication
  • Potentially broader spectrum activity
  • Examples: Plitidepsin (eEF1A inhibitor), Rintatolimod (TLR3 agonist)

Pan-Coronavirus Inhibitors:

  • Agents targeting conserved viral proteins
  • Potential for prophylaxis and treatment of future coronavirus variants
  • Examples: GC376 derivatives, calpain inhibitors

Economic Considerations and Stewardship

Cost-Effectiveness Analysis

Remdesivir:

  • High acquisition cost ($3,120 per course)
  • Potential savings through reduced length of stay
  • Cost-effective in appropriate patient populations

Nirmatrelvir/Ritonavir:

  • Lower acquisition cost ($530 per course)
  • Greatest value in preventing hospitalization
  • Limited ICU cost-effectiveness data

Stewardship Principles:

  • Prioritize patients with highest likelihood of benefit
  • Avoid use in patients unlikely to respond (very late in illness)
  • Consider stopping criteria for clinical non-response
  • Regular review of treatment duration necessity

Quality Improvement Initiatives

Implementation Strategies:

  • Standardized order sets with built-in safety checks
  • Automated drug interaction screening
  • Regular multidisciplinary rounds including pharmacist input
  • Outcome tracking and feedback systems

Pearl: Successful antiviral stewardship requires integration of clinical decision support tools with real-time patient data and multidisciplinary expertise.


Practical Clinical Pearls and Oysters

Clinical Pearls

  1. Timing is Everything: Antiviral efficacy is inversely related to time from symptom onset. Implement rapid testing and treatment protocols.

  2. Drug Interaction Vigilance: Create ICU-specific interaction protocols for nirmatrelvir/ritonavir. Many interactions are manageable with dose adjustments rather than absolute contraindications.

  3. Renal Function Focus: Always assess eGFR before remdesivir initiation. The SBECD excipient can accumulate in renal impairment.

  4. Hepatotoxicity Monitoring: Remdesivir-associated hepatotoxicity can be subtle. Daily LFT monitoring is essential, particularly in the first 72 hours.

  5. Route of Administration: Oral antivirals require functioning GI tract. Consider nasogastric/jejunal tube administration if swallowing is impaired.

Clinical Oysters (Common Mistakes)

  1. The Late Starter: Initiating antivirals after day 10 of illness when viral replication has waned and inflammatory processes dominate.

  2. The Interaction Ignorer: Prescribing nirmatrelvir/ritonavir without comprehensive medication review, leading to serious drug interactions.

  3. The Dose Forgetter: Failing to adjust doses for renal impairment, particularly with nirmatrelvir/ritonavir.

  4. The Monitor Misser: Inadequate monitoring of hepatotoxicity with remdesivir, mistaking drug-induced liver injury for disease progression.

  5. The Crusher: Attempting to crush or divide nirmatrelvir/ritonavir tablets, which destroys the film coating and alters absorption.

Clinical Hacks

  1. The Interaction App: Use clinical decision support tools or apps with real-time interaction checking for complex ICU medication regimens.

  2. The Timing Tool: Implement automated EMR alerts for antiviral timing based on symptom onset or positive test results.

  3. The Monitoring Matrix: Create standardized monitoring schedules with automatic laboratory ordering for antiviral safety parameters.

  4. The Team Approach: Establish rapid response teams for antiviral decision-making, including critical care, infectious disease, and pharmacy expertise.

  5. The Documentation Detail: Document symptom onset time, contraindications considered, and monitoring plans to facilitate continuity of care.


Future Directions and Research Priorities

Emerging Therapeutic Targets

Next-Generation Nucleoside Analogs: Research focuses on agents with improved bioavailability, reduced toxicity, and broader spectrum activity. Compounds like obeldesivir and sofosbuvir analogs show promise for respiratory viral infections.

Host-Targeted Therapies: Targeting cellular mechanisms essential for viral replication may provide broader spectrum activity and reduced resistance potential. Current investigations include:

  • Cellular protease inhibitors
  • Autophagy modulators
  • Innate immune enhancers

Inhalation Delivery Systems: Direct pulmonary delivery of antivirals may achieve higher local concentrations while minimizing systemic exposure and toxicity. Nebulized formulations of existing agents are under investigation.

Clinical Research Priorities

Critical Care-Specific Studies:

  • Optimal dosing in organ dysfunction
  • Combination therapy strategies
  • Duration of treatment optimization
  • Resistance monitoring protocols

Pharmacokinetic Studies:

  • Drug disposition during ECMO
  • Continuous renal replacement therapy effects
  • Plasma exchange implications
  • Protein binding alterations in critical illness

Technology Integration

Artificial Intelligence Applications:

  • Predictive modeling for antiviral response
  • Automated drug interaction screening
  • Resistance prediction algorithms
  • Treatment optimization protocols

Point-of-Care Diagnostics:

  • Rapid viral load quantification
  • Resistance mutation detection
  • Therapeutic drug monitoring
  • Real-time susceptibility testing

Conclusion

The integration of emerging antivirals into critical care practice represents both an opportunity and a challenge for intensivists. While these agents offer the potential to improve outcomes for critically ill patients with viral infections, their successful implementation requires careful attention to patient selection, timing, drug interactions, and monitoring protocols.

The evolution from limited antiviral options to a diverse therapeutic armamentarium demands sophisticated clinical decision-making and multidisciplinary collaboration. Success depends on understanding not only the pharmacology and efficacy of individual agents but also their integration into complex critical care treatment algorithms.

As new antivirals continue to emerge and our understanding of optimal use evolves, intensivists must remain committed to evidence-based practice while maintaining flexibility to adapt protocols based on emerging data. The lessons learned from COVID-19 antiviral deployment provide a foundation for managing future viral threats in critical care settings.

The future of antiviral therapy in critical care lies not just in the development of more potent agents, but in the creation of intelligent, adaptive treatment systems that can rapidly deploy appropriate therapy while minimizing adverse effects and resistance development. This requires continued collaboration between critical care physicians, infectious disease specialists, pharmacists, and clinical researchers to optimize outcomes for our most vulnerable patients.


References

  1. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the Treatment of Covid-19 — Final Report. N Engl J Med. 2020;383(19):1813-1826. doi:10.1056/NEJMoa2007764

  2. Hammond J, Leister-Tebbe H, Gardner A, et al. Oral Nirmatrelvir for High-Risk, Nonhospitalized Adults with Covid-19. N Engl J Med. 2022;386(15):1397-1408. doi:10.1056/NEJMoa2118542

  3. Jayk Bernal A, Gomes da Silva MM, Musungaie DB, et al. Molnupiravir for Oral Treatment of Covid-19 in Nonhospitalized Patients. N Engl J Med. 2022;386(6):509-520. doi:10.1056/NEJMoa2116044

  4. Goldman JD, Lye DCB, Hui DS, et al. Remdesivir for 5 or 10 Days in Patients with Severe Covid-19. N Engl J Med. 2020;383(19):1827-1837. doi:10.1056/NEJMoa2015301

  5. Spinner CD, Gottlieb RL, Criner GJ, et al. Effect of Remdesivir vs Standard Care on Clinical Status at 11 Days in Patients With Moderate COVID-19: A Randomized Clinical Trial. JAMA. 2020;324(11):1048-1057. doi:10.1001/jama.2020.16349

  6. Gottlieb RL, Vaca CE, Paredes R, et al. Early Remdesivir to Prevent Progression to Severe Covid-19 in Outpatients. N Engl J Med. 2022;386(4):305-315. doi:10.1056/NEJMoa2116846

  7. Aggarwal NR, Beaty LE, Bennett TD, et al. Real-world evidence of the neutralizing monoclonal antibody bebtelovimab in patients hospitalized with COVID-19. NPJ Vaccines. 2023;8(1):21. doi:10.1038/s41541-023-00621-6

  8. Zhou S, Hill CS, Sarkar S, et al. β-d-N4-hydroxycytidine inhibits SARS-CoV-2 through lethal mutagenesis but is also mutagenic to mammalian cells. J Infect Dis. 2021;224(3):415-419. doi:10.1093/infdis/jiab247

  9. Cao Z, Gao W, Bao H, et al. VV116 versus Nirmatrelvir-Ritonavir for Oral Treatment of Covid-19. N Engl J Med. 2023;388(5):406-417. doi:10.1056/NEJMoa2208775

  10. Stevens LJ, Pruijssers AJ, Lee HW, et al. Mutations in the SARS-CoV-2 RNA-dependent RNA polymerase confer resistance to remdesivir by distinct mechanisms. Sci Transl Med. 2022;14(634):eabo0718. doi:10.1126/scitranslmed.abo0718

  11. Iketani S, Mohri H, Culbertson B, et al. Multiple pathways for SARS-CoV-2 resistance to nirmatrelvir. Nature. 2023;613(7944):558-564. doi:10.1038/s41586-022-05514-2

  12. Tempestilli M, Caputi P, Avataneo V, et al. Pharmacokinetics of remdesivir and GS-441524 in two critically ill patients who recovered from COVID-19. J Antimicrob Chemother. 2020;75(10):2977-2980. doi:10.1093/jac/dkaa239

  13. Carothers C, Birrer K, Vo M. Acetaminophen for fever in critically ill patients with suspected infection. Am J Health Syst Pharm. 2021;78(23):2164-2168. doi:10.1093/ajhp/zxab298

  14. Ader F, Bouscambert-Duchamp M, Hites M, et al. Remdesivir plus standard of care versus standard of care alone for the treatment of patients admitted to hospital with COVID-19 (DisCoVeRy): a phase 3, randomised, controlled, open-label trial. Lancet Infect Dis. 2022;22(2):209-221. doi:10.1016/S1473-3099(21)00485-0

  15. WHO Solidarity Trial Consortium. Repurposed Antiviral Drugs for Covid-19 — Interim WHO Solidarity Trial Results. N Engl J Med. 2021;384(6):497-511. doi:10.1056/NEJMoa2023184

No comments:

Post a Comment

ICU-Related Infections: Beyond VAP and CLABSI

  ICU-Related Infections: Beyond VAP and CLABSI Ventilator-Associated Tracheobronchitis, Multidrug-Resistant Colonization, and Prevention S...